You are on page 1of 21

Review

published: 16 April 2018


doi: 10.3389/fimmu.2018.00648

Debunking the Myth of Exercise-


Induced Immune Suppression:
Redefining the Impact of Exercise
on Immunological Health Across
the Lifespan
John P. Campbell* and James E. Turner*

Department for Health, University of Bath, Bath, United Kingdom

Epidemiological evidence indicates that regular physical activity and/or frequent


structured exercise reduces the incidence of many chronic diseases in older age,
including communicable diseases such as viral and bacterial infections, as well as non-
communicable diseases such as cancer and chronic inflammatory disorders. Despite
Edited by:
the apparent health benefits achieved by leading an active lifestyle, which imply that
Annemieke Boots,
University Medical Center regular physical activity and frequent exercise enhance immune competency and regu-
Groningen, Netherlands lation, the effect of a single bout of exercise on immune function remains a controversial
Reviewed by: topic. Indeed, to this day, it is perceived by many that a vigorous bout of exercise can
Ana Maria Teixeira,
University of Coimbra, Portugal
temporarily suppress immune function. In the first part of this review, we deconstruct
Emily C. LaVoy, the key pillars which lay the foundation to this theory—referred to as the “open window”
University of Houston,
hypothesis—and highlight that: (i) limited reliable evidence exists to support the claim
United States
that vigorous exercise heightens risk of opportunistic infections; (ii) purported changes
*Correspondence:
John P. Campbell to mucosal immunity, namely salivary IgA levels, after exercise do not signpost a period
j.campbell@bath.ac.uk; of immune suppression; and (iii) the dramatic reductions to lymphocyte numbers and
James E. Turner
j.e.turner@bath.ac.uk
function 1–2  h after exercise reflects a transient and time-dependent redistribution
of immune cells to peripheral tissues, resulting in a heightened state of immune sur-
Specialty section: veillance and immune regulation, as opposed to immune suppression. In the second
This article was submitted
part of this review, we provide evidence that frequent exercise enhances—rather than
to Inflammation,
a section of the journal suppresses—immune competency, and highlight key findings from human vaccination
Frontiers in Immunology studies which show heightened responses to bacterial and viral antigens following bouts
Received: 19 November 2017 of exercise. Finally, in the third part of this review, we highlight that regular physical
Accepted: 15 March 2018
Published: 16 April 2018
activity and frequent exercise might limit or delay aging of the immune system, providing
Citation:
further evidence that exercise is beneficial for immunological health. In summary, the
Campbell JP and Turner JE (2018) over-arching aim of this review is to rebalance opinion over the perceived relationships
Debunking the Myth of Exercise- between exercise and immune function. We emphasize that it is a misconception to label
Induced Immune Suppression:
Redefining the Impact of Exercise any form of acute exercise as immunosuppressive, and, instead, exercise most likely
on Immunological Health improves immune competency across the lifespan.
Across the Lifespan.
Front. Immunol. 9:648. Keywords: exercise, physical activity, upper respiratory tract infections, open window hypothesis, infection
doi: 10.3389/fimmu.2018.00648 susceptibility, ageing, immunosenescence, immune competency

Frontiers in Immunology  |  www.frontiersin.org 1 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

INTRODUCTION the 1980s and 1990s, reviewed extensively elsewhere (7, 8, 14).
Findings from these early studies led to three principles of exer-
Lifelong physical activity1 is a potent means of reducing the cise immunology being formed, which have, until now, generally
risk of non-communicable diseases including cancer, car- been unchallenged in the literature: (i) infection risk is increased
diovascular disease, and other chronic inflammatory disorders after an acute bout of prolonged and vigorous aerobic exercise;
(1). Evidence also shows that a physically active lifestyle (ii) acute bouts of vigorous exercise can lead to a temporary
diminishes the risk of contracting a range of communicable reduction to salivary IgA levels culminating in a higher risk
diseases including viral and bacterial infections (2–6). In of opportunistic infections; and (iii) transient decreases in the
contrast to the widely accepted long-term health benefits that number of peripheral blood immune cells, which occurs in the
are achieved by regular physical activity, which imply that hours following vigorous exercise, represents a period of immune
immune competency and regulation are improved by frequent suppression. Over the years, these collective observations have
exercise bouts, the effect of a single bout of exercise on immune coalesced and led to the creation of the so-called “open-window”
function remains hotly disputed. Undeniably, acute vigorous hypothesis, which purports that the immune system is compro-
exercise has a profound effect on the phenotypic makeup and mised in the hours after vigorous exercise, leading to an increased
functional capacity of the immune system. Indeed, the behav- risk of opportunistic infections in the days thereafter. To this
ior of almost all immune cell populations in the bloodstream is day, the “open window” hypothesis continues to be discussed
altered in some way during and after exercise (7, 8). However, (15), despite the existence of contradictory evidence. Here, in
for decades, it has been widely accepted that these changes the first part of this review, we aim to dispel the “open-window”
result in a temporary decline in immune competency in the hypothesis by revisiting key research studies and highlighting
hours following exercise. In the first part of this review, we that limited evidence exists to support each of the three pillars
re-interpret these data and strive to dispel the misconception that lay its foundation.
that an acute bout of exercise is detrimental to immunological
health. In the second part of this article, we demonstrate that Exercise and Opportunistic Infections
rather than suppressing immunity, contemporary evidence It has been speculated for over a century that participation
shows that an acute bout of exercise improves immune sur- in physical activity heightens the risk of opportunistic infec­
veillance, for example leading to enhanced antibacterial and tions (16). However, deeper investigation into the relationship
antiviral immunity. Finally, in the third part of this article, we between exercise and infection susceptibility did not take place
summarize recent data suggesting that regular physical activity until the end of the twentieth century. At this juncture, the
and frequent exercise, which reduces systemic inflammatory principle focus of many of these studies in the late 1980s and
activity and improves aspects of immune function, also leads to early 1990s was to determine whether infection incidence was
alterations in classical biomarkers of an aging immune system. increased in elite and recreational athletes in the weeks fol-
These changes could be interpreted as limiting or delaying lowing mass participation distance running events. One of the
immunological aging (9–13). first studies from this era found that one third of 150 runners
participating in the 1982 Two Oceans 56 km ultramarathon in
PART A: IS IT TIME TO CLOSE THE Cape Town South Africa self-reported symptoms of upper res-
SHUTTERS ON THE “OPEN-WINDOW” piratory tract infections (URTIs; symptoms = runny nose, sore
throat, sneezing) within 2  weeks of the race (17). The control
HYPOTHESIS? A BOUT OF EXERCISE group, who were age matched and shared a home with another
DOES NOT SUPPRESS IMMUNE of the race competitors, reported only half the amount of URTIs
COMPETENCY in the same period (17). Similar observations were made in an
often-cited larger study of the 1987 Los Angeles Marathon (18).
A prevailing myth has formed in the literature that participating in Of 2,311 respondents who had completed the marathon and
an acute bout of aerobic exercise, particularly if it is vigorous and whom did not report an infection in the week prior to the race,
prolonged, can be detrimental to immune competency. The foun- 12.9% reported an infection in the week after the race compared
dations of this belief lay in research publications emanating from to only 2.2% of individuals who withdrew from the race for
reasons other than illness (odds ratio of infection in runners
versus non-runners = 5.9). A separate study conducted around
1 
“Physical activity” refers to activities undertaken during leisure time, at home, the same time found that shorter duration running events, such
as part of employment, or for transport purposes. “Exercise” is a component of as 5, 10 km, and half-marathons (21 km) did not appear to elicit
physical activity within the leisure time domain and refers to physical activities that
are planned, structured, repetitive, and undertaken for the purpose of improving or
an increased incidence of self-reported URTIs (19), thereby
maintaining components of physical fitness and/or sporting performance. When suggesting that URTI symptoms are increased only when the
individuals are referred to as being “active” or “inactive,” the description infers that duration of the exercise is long.
these people undertake (or fail to undertake) a defined level of exercise or physical A fundamental limitation of each of the aforementioned
activity (e.g., age-specific physical activity recommendations, such as those pub- studies is that none of the self-reported infections were clinically
lished by the World Health Organisation). In this review, the term “exercise” will
generally be used to describe the effects that active behaviours have on immune
confirmed by laboratory analyses (e.g., molecular or microbio-
competency. Individuals described as being “sedentary” accumulate prolonged logical techniques, such as polymerase chain reaction or bacte-
periods of behaviour eliciting low energy expenditure (e.g. sitting and lying). rial cultures). As a consequence, it was questioned whether the

Frontiers in Immunology  |  www.frontiersin.org 2 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

self-reported URTIs in these studies represented genuine infec- play a greater role in infection risk than exercise participation
tions. Clarifying this issue, a study employing nasopharyngeal per se. For example, recently, it has been demonstrated that air
and throat swabs in athletes who reported URTI symptoms over travel is a significant predictor of illness symptoms in athletes
a 5-month period—including periods of competition—found (32). Infections linked to air travel are exacerbated by long-haul
that few of the self-reported infections were of bacterial, viral, flights crossing multiple time zones, implicating many other fac-
chlamydial, or mycoplasmal nature (20). Indeed, of 37 episodes tors known to influence immune function, including exposure
of URTI reported by athletes in this study, only 11 of these to hypobaric hypoxia, radiation, temperature changes, sleep
(30%) had a positive laboratory diagnosis. These findings place disruption, fatigue, altered or inadequate diet, dehydration, and
the previously discussed marathon studies in a different light psychological stress (32–34).
(17, 18) and open the possibility that many of the URTIs reported Contrary to the aforementioned reports that exercise height-
were a symptom of other non-infectious causes. Indeed, of the ens infection incidence, it is often overlooked that other studies
of the non-infectious “URTIs” reported by Spence et  al. (20), indicate that exercise participation may in fact reduce the inci-
and likely captured elsewhere (17, 18), it is proposed that these dence of infections. For example, a recent prospective cohort
symptoms are a result of other causes, including allergy and study of 1,509 Swedish men and women aged 20–60 years found
asthma, non-specific mucosal inflammation, or airway epithe- that higher physical activity levels were associated with a lower
lial cell trauma due to increased ventilation or exposure to cold incidence of self-reported URTIs (35). A much smaller but
air (21). In the few cases of clinically confirmed URTIs, these very detailed analysis of illness records kept by 11 elite endur-
appear to be from viruses—in particular rhinoviruses (i.e., the ance athletes over a period of 3–16 years showed that the total
“common cold”)—rather than bacterial infection (20, 22), which number of training hours per year was inversely correlated with
is in line with the typical incidence and etiology of infections at sickness days reported (36). Similarly, another study of swim-
the population level (23). mers monitored for 4  years found that national level athletes
In the previously discussed clinical laboratory study of infec- had higher incidence of infections than more elite international
tion incidence in athletes (20), it is notable that the proportion of level athletes (37). Finally, studies of ultramarathon runners,
athletes with a confirmed infection during the 5-month period who undertake the largest volume of exercise among athletes,
was as follows: 2/20 controls (10%), 3/31 (10%) recreational have shown that these individuals report fewer days missed
athletes, and 6/32 (19%) elite athletes. Although this study was from school or work due to illness compared to the general
small, these data appear to align with observations from earlier population. For example, the mean number of sickness days
self-report studies which found that the incidence of URTI symp- reported over 12 months was 1.5 days in a study of 1,212 ultra-
toms was higher in those with the fastest race time and those who marathon runners and 2.8 days in a study of 489 ultramarathon
had completed a greater training volume pre-race (17, 18). These runners (38, 39). These studies compared their findings to data
early observations contributed to the formulation of the “J-shaped from the United States Department of Health and Human
curve” (24). This hypothesis infers that those who undertake an Services report in 2009, showing that the general population
excessive volume of exercise, over a period of weeks and months, report on average 4.4 illness days each year. Thus, a number of
sometimes referred to as “over-training” or “intensified training” studies challenge the “J-shaped curve,” indicating that athletes
(25, 26), are at a greater risk of infections (24). In this scenario, undertaking the largest training loads, become ill less frequently
other factors present prior to an acute bout of exercise, such as than athletes competing at, and training at, a lower level. These
psychological stress and anxiety (27–29), or nutritional deficien- findings have previously been conceptualized by extending the
cies (30) which are known to impact immune regulation, are “J-shaped curve” into an “S-shaped curve,” thereby suggesting
likely to impact immune competency and contribute to the risk that very elite athletes are better adapted to the demands of
of genuine URTIs, rather than the acute and transient immune their training (40). Given the nature of their design, very few
changes that arise after the acute bout of exercise itself; these acute of these reports—akin to many of the aforementioned studies
immunological changes arising after acute exercise are discussed showing increased infection risk among athletes following mass
later in this article (see Part A: “Is it Time to Close the Shutters on participation endurance events—used appropriate laboratory
the “Open-Window” Hypothesis? A Bout of Exercise Does Not diagnostics to confirm an infection. However, despite their
Suppress Immune Competency”; and see “Exercise and Salivary limitations, it is important to highlight that there are as many
IgA and Changes to Lymphocyte Frequency and Functional epidemiological studies showing that regular exercise reduces
Capacity in the Hours After Acute Exercise”). infections as there are studies showing exercise increases infec-
Moreover, we contend that attendance at any mass participa- tions, and that these studies are often overlooked in the exercise
tion event—whether it is a marathon or otherwise—is likely to immunology literature.
increase the risk of acquiring novel infectious pathogens, which It should also be considered whether the commonly reported
are in abundance due to the mass gathering of people. For exam- “increased frequency” of illness symptoms among athletic
ple, it has been shown that around 40% of individuals attending populations or those taking part in sporting events is indeed
the Hajj—a crowded religious event in Saudi Arabia—self-report more frequent than among the general population. For example,
an URTI (31). In this study, there was a greater risk of infection large studies have reported that approximately 7–10% of athletes
among those with the longest exposure to crowds (31). Thus, it competing in the Olympic Games report symptoms of illness
is important to consider that other underlying factors, often not during the competition weeks (41, 42). However, accumulating
measured in the context of exercise and illness studies, likely evidence suggests the incidence of infection among athletes is

Frontiers in Immunology  |  www.frontiersin.org 3 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

not substantially different to other populations. For example, in infection or immune impairment is confirmed, their trigger is
a telephone survey of 2011 adults considered to represent the more likely to be the physical, nutritional, and psychological
general population in the USA, 24% experienced a cold during wellbeing of the individual prior to undertaking the single bout
a 4-week period, which is a similar timeframe to many interna- of acute vigorous exercise. In the context of mass participation
tional sporting competitions (43). In another telephone survey of sporting events, it is likely that increased exposure to pathogens,
4,051 adults in the USA, 72% experienced at least 1 non-influenza or the influence of environmental factors that can affect immune
related URTI over 12  months, and on average, experienced 2 function (e.g., travel, sleep disruption) most likely explain genu-
infections annually (44). In a year-long Internet-based monitor- ine infections. Thus, we conclude that it is unlikely that vigorous
ing study of 627 individuals over 14  years of age in Germany, and prolonged exercise heighten the risk of infections and should
weekly acute respiratory illness rates were 2.7–8.2%, manifesting not be considered a deterrent to those seeking to become more
in 1.3–3.2 episodes annually (45). Thus, evidence suggests that the physically active.
frequency of illness episodes in athletic communities is similar to
the general population annually. Exercise and Salivary IgA
In the context of exercise participation in older age, it would A second mainstay of exercise immunology that has received
appear to be counterintuitive that the incidence of URTI symp- considerable attention over the last three decades is the assess-
toms appears to be inversely correlated with age: it has been ment of exercise-induced changes to mucosal immunity,
shown that URTI symptoms are more common in younger rather principally via measurement of IgA levels in saliva (52). Given
than older runners (18). While, once more, this aforementioned that IgA is the most abundant immunoglobulin in mucosal
study did not confirm infections by laboratory analyses, it is secretions and that its principle role is the inhibition of invading
notable that if acute exercise does suppress immune compe- pathogens, isolated changes to salivary IgA following exercise
tency, it might be expected that older adults—whom typically has been considered of some importance in light of the purport-
have inferior immune function—would be at greatest risk of edly higher risk of infections among athletes (7, 8).
exercise-induced immune suppression. Rather than exercise One of the earliest and most cited papers in this research area
per se, again, a more likely explanation for differences in illness found that IgA was reduced by 20% after 2–3 h of cross-country
symptoms between groups of athletes are other factors present skiing (53). Another study found that this effect is transient,
before competing, such as fatigue, nutritional deficiency, whereby salivary IgA concentrations decreased immediately
psychological stress, or environmental exposures. On the other after 2 h of intensive cycling exercise, and remained low in sam-
hand, proponents of the “open-window” hypothesis could ples collected 1  h post-exercise, but returned to normal levels
portend that experienced athletes have higher tolerance of the within 24  h (54). A criticism of these studies at that time was
symptoms associated with URTI, and/or have developed coping that the absolute IgA levels reported did not adequately control
methods or strategies to reduce symptoms. Alternatively, expe- for the amount of saliva produced, and thus these results may
rienced athletes may have evolved strategies to reduce infec- misrepresent IgA secretion. Although some studies measuring
tion risk by adopting good practice (e.g., sleep, diet, hygiene) IgA secretory rate (IgA protein concentration multiplied by
before, during and following attendance at a mass participa­ saliva flow rate) support the early findings with IgA concentra-
tion event. tion, others have shown profoundly contradictory results. For
Separately, it has also been questioned whether illness example, in alignment with prior observations, it was found in
symptoms—even if confirmed to be infectious—are a result of trained runners that IgA secretion rate decreased by 25% from
encountering a novel pathogen. For example, over a decade ago, pre-marathon to 90 min post-marathon (55). Likewise, in a sepa-
some studies suggested that reactivation of latent viruses—such as rate study, a 20% reduction in IgA secretion rate was observed
Epstein Barr Virus which had most likely infected the host during in elite athletes after a 2-h rowing exercise session (56). Several
childhood—was responsible for illness symptoms after exercise other studies of similar design reported analogous findings
(46, 47). Although these studies suggest new pathogens were (57–59); however, contradictory findings are also in abundance
not to blame, it was interpreted that exercise-induced immune but are much less cited in the literature. Indeed, an elegant study
suppression had resulted in loss of viral control. However, herpes exploring the effects of different exercise intensities, including
viruses can reactivate even with a fully functioning immune moderate- and high-intensity exercise to exhaustion, found that
system, for example, in response to adrenergic activity, reactive although saliva flow rate decreased, IgA secretion rate actu-
oxygen species and inflammatory cytokines (48–50), all of which ally increased in response to both of the exercise bouts. In the
increase during exercise. Moreover, recent evidence appears words of the authors, exercise to exhaustion has an “effect on the
to discount herpes virus involvement in causing symptoms of quantity of saliva, but not the quality of saliva” (60). Many other
illness, by showing that individuals previously infected with studies have also reported that exercise does not elicit a decrease
Cytomegalovirus, or those infected with both Epstein Barr Virus in IgA secretion rates following exercise (61–66).
and Cytomegalovirus, exhibited a lower incidence of illness symp- Any subtle isolated changes to IgA that occur after exercise
toms than individuals not latently infected (51). appear to be clinically insignificant as it would appear that the
Taken together, evidence that participation in an acute bout of increased incidence of URTI symptoms that has been purported
vigorous exercise leads to heightened infection incidence remains following vigorous and prolonged exercise is unrelated to salivary
spurious. If symptoms of URTI are observed after a bout of vigor- immunoglobulin status. A longitudinal field study of participants
ous exercise, the cause is unlikely to be infectious. However, if in the Comrades Marathon (86.5  km ultramarathon) in South

Frontiers in Immunology  |  www.frontiersin.org 4 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

Africa found that salivary IgA levels in the 4 weeks prior to the race, extensively reviewed elsewhere (78). Given these many consid-
and 2 weeks following the race, were unrelated to the incidence of erations, we propose that longitudinal measurement of salivary
self-reported URTI (67). In the aforementioned study, it was found IgA, as an isolated measure of immune competency within a
that symptoms of URTI were highest 4 weeks prior to the marathon, single host, and even more so between persons, depicts too
and URTI symptoms seemed to re-appear within many of the same confusing a picture, and it is ambitious to say that any subtle
athletes 1 or 2  weeks post-marathon. This study did not confirm changes to salivary IgA following exercise reflects immune
the re-emergence of infections by laboratory testing, but if this was suppression and a heightened risk of opportunistic infections.
indeed demonstrated in future studies, it again shows that acute Given the limitations of salivary IgA measurement, research is
exercise participation per se does not heighten risk of opportunistic being undertaken to explore mucosal IgA in other biofluids, and
infections. In this case, an underlying infection, not resolved prior a recent study has shown links between reduced tear IgA levels
to exercise participation, or some other idiosyncrasy, is perhaps to and infection incidence (79). Others have moved toward more
blame. Such conclusions appear to be supported by evidence from comprehensive oral immunity panels (80), and such strategies
athletes who report the most frequent illness symptoms. For exam- could benefit further from an integrative approach that, in addi-
ple, these individuals exhibit mostly anti-inflammatory cytokine tion to immune parameters, incorporates full dental examina-
responses when whole blood, collected at rest, is cultured ex vivo tion, oral inflammation biomarkers, and host mucosal ecology.
with antigens from diphtheria, tetanus, acellular pertussis, poliomy-
elitis, and hemophilus influenza type b (68, 69). These findings sug- Changes to Lymphocyte Frequency
gest the immune system may be functionally altered by underlying
and Functional Capacity in the Hours
illness, or is already different in these “illness-prone” athletes prior
to infection, rather than exercise affecting immune function per se. After Acute Exercise
A flaw in studies investigating the link between mucosal One of the most reproduced findings in human exercise physiol-
immunity and purported exercise-induced infection risk is that ogy is the profound and transient time-dependent change that
oral health status is rarely adequately evaluated. Salivary IgA is arises to the phenotypic composition and functional capacity
heavily involved in host-bacterial ecology and mucosal homeo- of lymphocytes in the peripheral bloodstream in response to
stasis (70, 71). As optimal oral health is rare in adults—with a single bout of exercise (8). During vigorous aerobic exercise,
nearly all exhibiting caries, gingivitis or periodontitis—pro- it is commonly observed that peripheral blood lymphocyte
found between-person IgA variation has been reported, which frequency—and, concomitantly, the functional capacity of the
is dependent on oral health status (71). Moreover, periodontal lymphocyte pool—is dramatically increased, leading to the
diseases are complex and multifactorial, and as a result, studies concept that, during exercise, exercise appears to “stimulate”
report large fluctuations in IgA levels relative to disease status the immune system. On the other hand, in the hours follow-
between persons, probably due to the bespoke ecological makeup ing exercise, it is typically observed that total peripheral blood
of different host mucosa (71). In addition, oral disease is a com- lymphocyte frequency—and the functional capacity of the lym-
mon problem in athlete populations (72), which is likely to be phocyte pool—is decreased below pre-exercise levels, leading
caused by high volume and frequent carbohydrate consumption, some to propose that exercise induces a short-term window of
and in some, a neglect of oral hygiene, perhaps due to practical immune suppression (termed the “open-window” hypothesis).
constraints. Thus, changes to oral inflammatory status has not The purpose of this part of our review is to outline that it is
been adequately considered, and emerging salivary biomarkers a misconception to state that the “reductions” to lymphocyte
of oral inflammation, such as immunoglobulin-free light chains frequency and function, that arise in the hours following acute
(73), may offer a means of controlling for this confounder. As exercise, reflects immune suppression, and instead we emphasize
highlighted elsewhere (70), salivary IgA is also highly vulnerable that during this post-exercise period, the immune system is in a
to short-term variation, in particular, due to circadian rhythms, heightened state of immune surveillance and regulation.
typically peaking in the morning, and falling thereafter (74).
As salivary IgA secretion is controlled by the parasympathetic Transient Changes to Blood Lymphocyte
and sympathetic nervous system, psychological stress also plays Frequency in the Hours Following Exercise
a powerful role in regulating IgA levels (75). Animal models The classic biphasic response of lymphocytes to acute steady
suggest that salivary IgA levels could vary up to 27-fold within state vigorous exercise lasting for around at least 45–60 min-
the same host over a short period of time (76). Salivary IgA utes, is first characterized by a dramatic lymphocytosis. This
levels are also affected by factors such as sex differences (77), response is typified by a dramatic influx of natural killer cells,
diet, ethnicity, disease, medications, tobacco, and phase of which rise by up to 10-fold, and CD8+ T cells which increase to
the menstrual cycle, as reviewed elsewhere (71). To overcome a lesser—but still profound—extent by approximately 2.5-fold
some of these variations in salivary IgA, it is often the case that (81). This exercise intensity-dependent mobilization is driven
studies evaluate only secretory IgA (sIgA; i.e., IgA containing in part by increased shear forces and blood pressure during
the secretory component) as this represents IgA produced by exercise causing a non-specific flushing of the marginal pools
local mucosal plasma cells, and not IgA from the bloodstream (82) but, moreover, is principally governed by adrenergic
transported via crevicular fluid. While this approach may stimulation of beta-2-adrenergic receptors on the surface of
reduce some confounding error, most IgA in saliva contains the lymphocytes, arising from adrenaline released during exercise,
secretory component and is, itself, subject to large variation; causing endothelial detachment and subsequent recirculation

Frontiers in Immunology  |  www.frontiersin.org 5 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

of lymphocytes into the bloodstream (83–85). Indeed, the numbers post-exercise (89, 101, 102), apoptosis could be a small
lymphocyte mobilization response observed during exercise contributor to exercise-induced lymphopenia, but this process
appears to broadly mirror the differential expression of beta-2 of cell death is likely to be beneficial given the stimulation of
adrenergic receptors on lymphocytes: natural killer cells > progenitor cells from the bone marrow (95).
CD8+ T  cells  >  B  cells  >  CD4+ T  cells, including regulatory While it has not been shown in humans that exercise—in
T cells (81, 86–88). Upon exercise cessation, the classic biphasic line with rodent models—causes the redistribution of immune
exercise response is next characterized by a dramatic decrease in cells to peripheral tissues, further support for a coordinated,
the frequency of lymphocytes in the bloodstream. This nadir is exercise-induced immune surveillance response elicited
typically observed approximately 1–2 h post-exercise when the by lymphopenia, is revealed by studying the phenotypic
lymphocyte numerical count is lower than pre-exercise levels; charac­teristics of the cells that preferentially mobilize and
lymphocyte frequency normally returns to pre-exercise levels subsequently extravasate out of the circulation after exercise.
within 24  h (87, 89, 90). The lymphopenia that occurs 1–2  h With regard to natural killer cells—the most exercise-
later is exercise intensity dependent and the most profound responsive lymphocyte subset—CD56dim cells are preferen-
reductions during this period are typically observed among tially redeployed rather than their CD56bright counterparts
natural killer cells and CD8+ T  cells (90). Akin to purported (81). CD56dim cells are a mature subset of natural killer cells
reductions to salivary IgA, discussed earlier, it was perceived which have exclusive migratory potential for non-lymphoid
that the numerical decline of blood lymphocytes that arises dur- tissue and potent effector capabilities, including the capacity
ing this time represented “double jeopardy” (89), temporarily to produce high amounts of perforin and granzyme, whereas
exposing an individual to impaired immune competency and CD56bright cells are a more immature regulatory cell subset
concomitantly providing an “open-window” for opportunistic (103) and reside in secondary lymphoid organs, typified
infections (91, 92). by their cell-surface expression of CD62L and CCR7 (104).
Rather than suppressing immune competency however, a more CD56dim natural killer cells can be further dissected, into cells
contemporary viewpoint is that this acute and transient lym­ with highly potent effector function based on loss of NKG2A
phopenia 1–2 h after exercise is beneficial to immune surveillance and expression of killer immunoglobulin-like receptors and
and regulation. Indeed, in what appears to be a highly specialized CD57 (105, 106). In a recent study, it was shown that these
and systematic response, it is widely proposed that exercise rede- natural killer cells, which are capable of rapid effector func-
ploys immune cells to peripheral tissues (e.g., mucosal surfaces) tions, are preferentially redistributed after exercise (107, 108).
to conduct immune surveillance. Here, these immune cells are Synergistically, T cells also appear to exert heterogeneous but
thought to identify and eradicate other cells infected with patho- highly coordinated responses to acute exercise. Indeed, it is
gens, or those that have become damaged or malignant, termed consistently observed that discrete populations of CD8+ but
the acute stress/exercise immune-enhancement hypothesis (93). not CD4+ T cell subsets are redeployed by exercise. For some
A seminal study by Kruger and colleagues, using fluorescent cell time, there was confusion pertaining to the exact behavior
tracking in rodents, found that T  cells are redeployed in large of CD8+ T  cells in response to exercise. Rather than losing
numbers to peripheral tissues including the gut and lungs, and or gaining markers of adhesion or activation—as evaluated
to the bone marrow following exercise (84, 94). In line with elsewhere (8)—these changes represent a uniform redeploy-
Dhabhar’s theory, it is hypothesized that this redistribution ment of a preferentially mobilized group of memory cells. In a
reflects heightened immune surveillance in sites where patho- flurry of studies about a decade ago, it was shown that exercise
gens are likely to be encountered during and after exercise (i.e., selectively mobilizes memory CD8+ T cells with a phenotypic
lungs, gut). This response has also been proposed to maintain propensity for homing to peripheral tissues—typified for
immune homeostasis via augmented regulatory activities (12). example by CD11, and not CCR7 or CD62L expression—and
In this context, evidence implies that bone marrow homing and the distinctive capacity to mount rapid effector functions (81,
subsequent apoptosis of senescent T cells stimulates the produc- 109–111). This response presumably facilitates the detection
tion or mobilization of new progenitor cells into the periphery and elimination of neoplastic, stressed or infected cells in
(95), which has been hypothesized as an exercise-induced means synergy with natural killer cells, as proposed elsewhere (112).
of maintaining a younger immune system (12), discussed later Aligned with the immune surveillance theory of Burnet
in Part C “Does Exercise and Regular Physical Activity Influence and Thomas, reviewed elsewhere (113), these results imply
Immunological Ageing.” Links between exercise-induced that sentinel cells of the immune system are redeployed by
apoptosis and lymphopenia have in the past been interpreted as exercise-induced perturbations to stress hormones, to exert
detrimental, with speculation that apoptosis could be responsible effector functions against neoplastic, stressed, or infected cells
for the fall in lymphocyte number in the hours after exercise (96, in the hours following exercise. This process, which occurs
97). Other studies have reported increased lymphocyte apoptosis daily in a natural diurnal process (114), orchestrated subtly by
immediately after exercise (i.e., as a result of the large mobili- stress hormones (115, 116), appears to be primed in response
zation of cells) but not in the hours following exercise during to exercise, leading to enhanced immune surveillance (117).
lymphopenia (98–100). Although the magnitude of lymphocyte Principles of the acute stress/exercise immune-enhancement
apoptosis reported in studies is dependent on the measurement hypothesis continue to be investigated. For example, it has
technique, typically <10% of lymphocytes undergo post-exercise recently been shown that acute exercise does not preferentially
apoptosis (96, 97). Given the 30–60% decrease in lymphocyte mobilize CD8+ T cells and natural killer cells with the capacity

Frontiers in Immunology  |  www.frontiersin.org 6 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

for skin-homing (118). However, skin-homing in this context changes in overall cell function, indicated by the performance
is a role that may be fulfilled by exercise-responsive mucosal- of the sampled cells being assayed. For example, among CD8+
associated invariant T cells (119); but further work is needed T cells, subsets that exhibit strong effector function (e.g., CD45
in this area. RA+CD27−CD28−CCR7−CD62L−CD57+), substantially increase
A key example illustrating how exercise-induced immune during exercise (81, 123). Thus, during exercise, blood is pre-
cell redistribution is beneficial to host health can be found in dominantly occupied by cells capable of responding strongly
the rapidly emerging field of exercise oncology. Indeed, a recent (i.e., IFN-gamma production) to in vitro stimuli, and therefore,
seminal study demonstrated inhibition of tumor onset and dis- many studies have shown an increase in IFN-gamma production
ease progression across a range of tumor models in voluntarily by cells isolated close to the exercise stimulus. In the hours fol-
active rodents (112). In this work, natural killer cell infiltration lowing exercise, the same effector CD8+ T cells are subsequently
was significantly increased in tumors from active versus inactive redeployed to peripheral tissues, and, as such, this results in
rodents, leading to the conclusion that the presence of natural the blood having fewer cells capable of responding strongly
killer cells (but perhaps also T cells) in tumor sites, redeployed to in  vitro stimuli, thus explaining the commonly reported
by adrenaline during exercise stress, “provides a spark” for decrease in cellular function post-exercise. These effects have
tumor elimination, in what could be considered a form of been neatly demonstrated approximately two decades ago when
“exercise immunotherapy” (112, 120). Importantly, administra- it was shown that IFN-gamma production by stimulated CD8+
tion of propranolol—a beta 2 adrenergic blocker—abolished T cells is reduced 2 h after completing a prolonged 2.5 h bout
the adrenaline-induced redistribution of immune cells, and of cycling (124). Importantly, it was shown that this reduced
nullified the anti-tumor effect of exercise on neoplastic growth capacity to produce IFN-gamma was due to a reduced number
(112). While these studies are limited to rodents, there is grow- of IFN-gamma-positive CD8+ T cells in peripheral blood at the
ing evidence that exercise may promote anti-cancer effects in same time-point (124, 125).
humans. For example, in a key study recently conducted in The same principles apply to other cell functions, such as
humans, it was shown that natural killer cells with a highly in vitro proliferation in response to mitogenic stimuli. However,
mature effector phenotype are preferentially redistributed after with this measurement in particular, the commonly reported
exercise, and have the capacity to exert augmented cytotoxicity increase in T cell proliferation immediately after acute bouts of
against myeloma and lymphoma cells in vitro (107, 108). In light exercise is also strongly influenced by laboratory processes and
of these results, research is now being conducted to harness the in  vitro assay conditions (e.g., blood processing, temperature,
beneficial impact of acute exercise on lymphocyte kinetics for mitogen selection) (126). A recent meta-analysis of 24 studies
the purposes of cancer immunotherapy (121). It is beyond the concluded that lymphocyte proliferation is suppressed following
scope of this review to discuss other findings in this exciting field acute bouts of exercise, and that a greater magnitude of sup-
and we briefly conclude that the aforementioned studies imply pression is caused by exercise lasting longer than 1 h, regardless
that exercise-induced lymphocytosis, and the lymphopenia that of exercise intensity (127). However, this meta-analysis did
follows, is beneficial to the immune system’s capacity to iden- not examine the most important determinant of cell function
tify and neutralize damaged and neoplastic cells in peripheral following exercise: the time-dependent changes in the cellular
tissues. Furthermore, in the context of neoplastic growth, this composition of the samples assayed. Thus, findings such as these
process may be directly responsible for reduced incidence of should be interpreted with caution if analyses did not differentiate
cancer among physically active people across the lifespan (122). between studies collecting samples immediately after exercise or
Further comprehensive discussion of the role of exercise and in the hours following.
lymphocyte kinetics in anti-tumor responses can be found else- As with research focusing on T cells, a similar group of stud-
where (117). Clearly more research is needed in this area, and ies citing reductions to natural killer cell cytotoxicity following
a shift in focus toward investigating the benefits—rather than acute exercise, reviewed elsewhere (128), did not always take into
purported detrimental effects—of exercise on health, is no doubt account dramatic shifts in the constitutional makeup of the natu-
underway and will be a key focus for exercise immunologists in ral killer cell compartment, which is known to change in response
the coming years. to exercise (81). Once more, changes to the functional capacity of
the total natural killer cell pool are likely to have been misrepre-
Transient Changes to Blood Lymphocyte sented, given that many of these cells, with potent effector func-
Function in the Hours Following Exercise tions, are redistributed to peripheral tissues following exercise
A common misinterpretation, brought about by the exercise- cessation. The principles discussed herein regarding lymphocyte
induced reductions to blood lymphocyte frequency in the hours function are also broadly applicable to the assessment of function
following exercise, is the observation that the functional capacity in other cells, such as neutrophils and monocytes; the response
of immune cells in the peripheral blood is reduced in the hours of these cells to exercise is beyond the scope of this article and is
following vigorous exercise. As measurements of cell function reviewed elsewhere (8).
in peripheral blood are entirely dependent on the cells present Taken together, it is important to emphasize that statements
at the time of sampling, a change to the composition of the cells such as “acute intensive exercise elicits a depression of several
in blood in the hours after exercise—as outlined in the section; aspects of acquired immune function” and “prolonged bouts of
“Transient Changes to Blood Lymphocyte Frequency in the heavy exertion reduce the normal functioning of all major immune
Hours Following Exercise”—will consequently lead to parallel cell subtypes” mentioned elsewhere (8, 15) should be interpreted

Frontiers in Immunology  |  www.frontiersin.org 7 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

with caution. We conclude that the results of studies exploring Exercise and Immune Responses
the effects of acute exercise on cell function must be considered to Experimental In Vivo Challenge
very carefully in light of the time-dependent changes in the
cellular composition of blood that typically arise following a
Across the Lifespan
In a research context, the most clinically relevant model to
vigorous bout.
assess in  vivo challenge in a controlled manner is via vaccina-
Summary: Exercise Induces Lymphocyte tion. Vaccine administration assesses the integrated capacity of
Immune Surveillance Not Immune Suppression the immune system to recognize and process antigen, leading to
In summary, strong evidence implies that a reduction in the antigen neutralization. In a clinical research context, vaccination
frequency and function of lymphocytes (and other immune responses are principally quantified clinically in two ways, either
cells) in peripheral blood in the hours following vigorous via antibody production from antigen-specific plasma cells or via
and prolonged exercise does not reflect immune suppression. cytokine responses—typically IFN-gamma production—from
Instead, the observed lymphopenia represents a heightened T cells stimulated with cognate antigen.
state of immune surveillance and immune regulation driven by
a preferential mobilization of cells to peripheral tissues. As such,
Vaccination: Effects of a Single Exercise Bout
nutritional interventions, which have been employed to dampen
Evidence from an array of studies, evaluated recently in a com-
the magnitude of exercise lymphopenia (124, 129) are unlikely
prehensive review elsewhere (9), indicates that a single acute bout
to reduce the incidence of infections, but inter­ventions that
of exercise appears to enhance immune responses to vaccination
augment exercise-induced lymphocyte trafficking may provide
in both younger and older individuals. The majority of studies
benefits (130).
to date have examined muscle-damaging upper arm resistance
exercise performed close to the time of vaccination which is
PART B: REGULAR PHYSICAL ACTIVITY administered shortly after the regimen into exercised muscle.
AND FREQUENT EXERCISE AUGMENT However, other modes of exercise, including acute bouts of whole
ASPECTS OF IMMUNE COMPETENCY body aerobic activity, have also been investigated. Six of the eight
ACROSS THE LIFESPAN trials identified in the aforementioned review indicated a sta-
tistically significant exercise-induced enhancement of immune
Contrary to a commonly held belief—outlined in Part A “Is it responses against constituent antigens contained within the
Time to Close the Shutters on the “Open Window” Hypothesis? vaccine administered (133–138). It is notable that in five of these
A Bout of Exercise Does Not Suppress Immune Competency?”— studies, statistically significant benefits were found where the
that acute vigorous exercise can suppress aspects of immune vaccine strains appeared to have lower immunogenicity (9). For
function, an increasingly large body of research indicates example, it was shown in a trial of 133 young adults, approximately
that both single bouts of exercise, or frequent participation 20 years of age, receiving either a full- or half-dose Pneumovax-23
in regular exercise, can act as an adjuvant to stimulate the (a pneumococcal vaccine), that those who exercised at the time
immune system. Numerous methods exist to assess the effects of receiving the half-dose vaccine had heightened responses to
of behavioral interventions on immunity (131) but arguably five of the eleven pneumococcal strains contained in the vaccine,
the optimal means of evaluating global immune competency whereas no differences were observed for the other six strains;
at a systems level is via assessment of the immune response to and no benefits of exercise were observed for the full-dose vac-
in vivo challenge, ideally with a novel and clinically recognized cine (137). As such, given the potential effectiveness of exercise
pathogen, for example via vaccination (132). Thus, here, in the as an adjuvant in  situations where vaccine immunogenicity is
first section of Part B “Regular Physical Activity and Frequent low, studying the effects of exercise on antibody responses in
Exercise Augment Aspects of Immune Competency Across the older adults—whom typically exhibit impaired responses—has
Lifespan,” we focus on the influence that exercise has on immune received considerable attention. In one recent study, it was found
responses to antigenic challenge, which requires a coordinated that antibody responses in women 55–75  years of age, were
response from most, if not all, innate and adaptive immune significantly improved when moderate-intensity aerobic exercise
system components. In light of the age-associated decline in was performed immediately prior to vaccination; however, no
immune competence with aging—caused in part by underlying beneficial effects were found in men (138). Another trial reported
changes to the numerical, phenotypic, and functional capacity no benefits of a single 45-min bout of moderate-intensity walking
of almost all innate and adaptive immune cells—the second sec- exercise on the immune response to influenza and pneumococ-
tion of Part B “Regular Physical Activity and Frequent Exercise cal vaccination in women around 47 years of age (139). Finally,
Augment Aspects of Immune Competency Across the Lifespan,” a very recent study found no effect of a bout of resistance exer-
will evaluate the impact aging has on the immune benefits that cise on antibody responses to influenza vaccination in adults
can be attained from exercise throughout the lifespan. A detailed approximately 73 years of age (140). It is possible that a number
discussion of immunological aging processes and the influence of factors including immunological aging, biological sex and
that an active lifestyle has on established biomarkers of an variations in sex hormone levels, and perhaps latent infections
aging immune system is covered in Part C “Does exercise and (e.g., herpes viruses) (141–143) limit the immunostimulatory
Regular Physical Activity Influence Immunological Ageing?” of effects of exercise, and many studies have not adequately con-
this article. trolled for these factors. Alternatively, in light of the mechanisms

Frontiers in Immunology  |  www.frontiersin.org 8 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

proposed in the acute stress/exercise immune-enhancement (155). Thus, it may be the case that exercise enhances immune
hypothesis (93), it is plausible that more intensive exercise may responses, beyond those captured using maximal antibody titer
be needed to elicit enhanced vaccine responses. Despite null as an endpoint. In accordance with this view, an elegant rodent
findings, it is important to point out that few, if any, studies inves­ study conducted by an independent group (156) found that mice
tigating the effects of acute exercise on vaccination responses exercised for 20–30 min at moderate intensity 4 h after intrana­
have reported exercise-induced impairment to immune respon­ sal influenza exposure had a substantially higher survival rate
ses, and rather, these studies report that exercise has no effect, (18 of 22 survived; 82%) when compared to mice that did not
or in some cases a beneficial effect, on the immune response to exercise after influenza exposure (10 of 23 survived; 43%).
vaccination in older adults.
Contact Sensitivity Reactions and Acute
Vaccination: Effects of Frequent Exercise Bouts Exercise Bouts
Data from vaccine studies exploring the effects of regular physical More recent studies have examined the effects of acute exercise
activity or frequent exercise training on the immune response to on immune competency using other in  vivo models of immune
vaccination provides robust support for the argument that exercise challenge that, in principle, also assess the coordinated efforts of
enhances, rather than suppresses immunity. Indeed, at least eight immune system components. These studies have employed con-
studies have demonstrated heightened vaccination responses tact-sensitivity reactions by topically applying to skin, the dendritic
in older adults, typically over 60 years of age, who are physically cell and T  cell stimulant (or attractant) diphenylcyclopropenone
active (144–151). For example, an early study categorized adults (DPCP), and the non-specific inflammatory stimulant, croton oil
aged 62 years or older, into one of three groups: active (undertak- (157, 158). For example, in studies of young adults (approximately
ing at least 20 min of vigorous exercise three or more times per 20–30 years of age), by applying a primary sensitizing dose of DPCP
week), moderately active (undertaking regular exercise but with 20  min after 2  h of moderate-intensity treadmill running, and
lower intensity, frequency, and/or duration), or sedentary (non- assessing recall challenge 4 weeks later, it has been concluded that
exercisers). Two weeks after influenza vaccination, it was shown this form of exercise impairs both the induction of T cell immunity
that serum anti-influenza IgG and IgM titers were higher in and the memory response (159, 160). Thirty minutes of moder-
active versus sedentary adults, and so too were peripheral blood ate- or vigorous-intensity running had no effect, and no forms
mononuclear cell responses to antigen-specific stimulation (144). of exercise modulated the non-specific inflammatory challenge
In addition, a recent study has shown that men aged 65–85 years, in response to croton oil (159, 160). Although these findings are
who regularly undertook moderate or vigorous exercise training, biologically interesting, the clinical relevance of exercise-induced
exhibited higher antibody responses compared to inactive controls change is unclear, in part, because the process of DPCP-induced
in response to an influenza vaccine (151). Data linking habitual immune modulation is not well defined (158) unlike the immune
levels of physical activity to enhanced immune competency in response to antigen administration by vaccination.
humans are supported by evidence from animal studies, and Summary: Exercise Enhances Immune Responses
show that the immunological benefits of exercise may be particu- to In Vivo Antigenic Challenge
larly beneficial in enhancing otherwise poor responses in older We conclude that there is growing evidence from a powerful
age (152). array of studies in humans and rodents, indicating that exercise
enhances, or at least does not suppress immune responses
Interpreting Data From Vaccine Trials to in  vivo challenge in younger and older individuals. These
A major criticism of vaccine and exercise trials conducted in observations—which contradict those predicated by the “open-
humans is that many solely focus on the maximal antibody titer window” hypothesis—support the contention that an acute bout
following vaccination, and it is not practical to follow-up with of exercise has no detrimental immune consequences for health.
investigations into infection incidence as a gauge of protection Thus, exercise should be encouraged, particularly for older adults
status following vaccination (153). As such, it is unknown who are at greatest risk of infections and who may obtain the
whether differences observed with absolute antibody titers, or greatest exercise-induced benefits to immune competency; an
the amount of IFN-gamma produced from stimulated T  cells, overview of the impact of aging on the immunological benefits
between exercise and control groups, is representative of clini- that can be attained from exercise is described next.
cally meaningful benefits in terms of protection from infections.
Three elegant studies in rodents imply that benefits, beyond
antibody titer, may be brought about by acute exercise. In one of
Does Aging Influence the Immunological
these studies, it was found that antibody responses to influenza Benefits of Exercise and Regular Physical
exposure were lower in rodents that exercised around the time of Activity?
exposure, compared to those that did not exercise, yet, exercised Effect of a Single Exercise Bout
mice were still protected and did not exhibit any signs of infec- Research investigating the effects of exercise on immune
tion upon re-exposure to the virus (154). Moreover, in an earlier function has sought to establish whether the observed ben-
study by the same authors, it was found that mice undertaking an efits, as outlined earlier in young adults, such as exercise-induced
acute bout of exercise before being expose to influenza exhibited immune cell mobilisation, that has been implicated in protection
a lower severity of infection and had enhanced viral clearance against cancer, is also applicable to older adults. For example, it
and lower inflammation in the lungs in the days following has been reported that the magnitude of T  cell mobilization

Frontiers in Immunology  |  www.frontiersin.org 9 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

in response to acute vigorous exercise is smaller in older of age (168). Improvements in these common measurements
(65 ± 1 years) compared to younger (22 ± 1 years) adults (161, of immune competency, however, are not always consistent in
162). However, in this study, it was also shown that following longitudinal studies employing exercise training interventions,
exercise, the magnitude of T  cell proliferation in response to with around half of studies reporting improvements, and half
mitogens was smaller in young adults, whereas a similar exercise- reporting no change (163–165). One reason for this could be
induced stimulation of natural killer cell cytotoxicity was obser­ because the dramatic effects of Cytomegalovirus on driving
ved for both groups (161, 162). It is beyond the scope of this immunological aging was not considered by most of these studies,
review to fully critique investigations examining the influence of and it is feasible that results would be different when examining
single exercise bouts on the function of different immune cells, individuals who are latently infected compared to those who are
with comparisons made between younger and older individuals seronegative. Importantly however, no studies report impaired
across the lifespan; we refer the reader to comprehensive reviews immune competency from increased participation in structured
on this topic (163–165). Nevertheless, it is important to point exercise. Altogether, we conclude that despite declines in fitness
out that many studies in this area are difficult to interpret: at the and immune competency, aging does not appear to negate the
time of their publication, the influence of Cytomegalovirus infec- immunological benefits that can be attained from exercise, and
tion on the magnitude of exercise-induced immune responses indeed, frequent participation in exercise across the lifespan may
was not known and was therefore not considered (123, 166). lead to immune benefits, even in older age.
Moreover, while the magnitude of change to lymphocyte kinet-
ics is likely to be important for detecting and eliminating viral
and bacterial infections and neoplastic cells, this process is
PART C: DOES EXERCISE AND REGULAR
complex to study, and comparisons between younger and older PHYSICAL ACTIVITY INFLUENCE
people is difficult, partly due to other age-associated changes IMMUNOLOGICAL AGEING?
that influence the physiological response to exercise, such as
the decline in fitness (e.g., sarcopenia, cardiorespiratory fitness) Since the first exercise immunology research in the early 1900s,
with age. It is likely that some, or all of these factors impact and the substantial increase in scientific interest from the late
upon on the efficacy of exercise as an adjuvant to vaccination 1980s and early 1990s (169), studies examining interaction
responses in older adults, outlined earlier in the section “Exercise between immune function and lifestyle factors such as exercise and
and Immune Responses to Experimental In  Vivo Challenge physical activity have become common. Although a few exercise
Across the Lifespan.” In light of the challenge interpreting the studies published in the last 10–15 years have investigated some
clinical relevance of the aforementioned lymphocyte kinetics immunological processes relevant to aging, health, and disease,
studies, from here onward, we briefly evaluate the impact of the theme of exercise-induced “immune suppression” continues
regular physical activity or frequent exercise bouts on immune to influence the design of new studies or at least features in the
competency across the lifespan, using measurements in samples interpretation of findings and is often justified or contextualized
collected from participants at rest. with relevance to self-reported illness symptoms among athletes.
As outlined in Part A “Is it Time to Close the Shutters on the
Frequent Bouts of Exercise “Open-Window” Hypothesis? A Bout of Exercise Does Not
Immune competency at rest has been assessed in cross-sectional Suppress Immune Competency,” there is limited reliable evidence
studies, comparing elderly individuals differentiated by physi- to show that exercise heightens risk of opportunistic infections,
cal activity level or cardiorespiratory fitness, or by examining but there is, however, a growing body of evidence to show that
immune function before and after structured exercise training exercise enhances, rather than suppresses, immune competency,
interventions. For example, it has commonly been reported as summarized in Part B: “Regular Physical Activity and Frequent
among the elderly, that the most active participants, compared to Exercise Augment Aspects of Immune Competency Across the
those who are least active, show the highest T cell proliferation and Lifespan.” The beneficial effects of exercise on immune function
cytokine production in response to mitogens (163–165). Fewer are likely to be greatest for elderly people exhibiting the age-
studies have assessed innate immune competency, but higher associated deterioration of immune competency, also referred
natural killer cell cytotoxicity has been consistently shown among to as immunosenescence. Moreover, preliminary evidence sug-
the elderly who are active compared to less active age-matched gests that physical activity and regular structured exercise may
controls (163–165). Recent studies have expanded measurements even limit or delay immunological aging. Here, in Part C: “Does
into other innate immune cells such as neutrophils. For example, Exercise and Regular Physical Activity Influence Immunological
a recent cross-sectional study of 211 elderly adults, showed that Ageing?” we evaluate whether an active lifestyle influences
neutrophils from the 20 most active participants, compared to the immunosenescence.
20 least active participants, migrated toward interleukin (IL)-8
with greater chemotactic accuracy, but there were no differences The Aging Immune System
in chemotactic speed (167). In addition, a recent exercise train- Aging is associated with profound changes to the numerical, phe-
ing study has shown in both young and middle-aged adults, that notypic, and functional capacity of almost all innate and adaptive
10  weeks of moderate-intensity continuous cycling training, immune cells, resulting in altered immune responses. Some innate
or high-intensity interval cycling training, improve neutrophil immune cells exhibit numerical, phenotypic, and functional altera-
and monocyte phagocytosis and oxidative burst irrespective tions with aging, whereas others appear to be less affected (170).

Frontiers in Immunology  |  www.frontiersin.org 10 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

For example, the numbers and functions of eosinophils, basophils, exhibiting similar changes to their phenotype, functional proper-
and mast cells appear to be largely unchanged with aging, or at ties, and specificities as with the broader population of CD4+ and
least, there is not a clear effect of age based on the limited current CD8+ T cells (171). There is an age-associated decline in the total
literature (170). Neutrophil numbers often increase with aging, but number of γδ T cells; however age per se, in the absence of chronic
these cells exhibit diminished phagocytosis and impaired chemo- infections, is associated with a decline in Vδ2 cells (60–80% of γδ
taxis, although chemokinesis is maintained (170). Natural killer T cells), whereas Vδ1 (15–20% γδ T cells) remain stable (188).
cells increase with aging, driven by an accumulation of cytotoxic Some evidence suggests that γδ T  cell proliferative responses
CD56dim cells but a decline in regulatory CD56bright cells, and over- are impaired with aging, perhaps due to increased susceptibility
all, cytokine production and cytotoxicity are less on a per cell basis of Vδ2 cells to apoptosis (189, 190). Natural regulatory T  cells
(170). Other innate lymphocytes, such as invariant CD1d-restricted increase with aging whereas inducible regulatory T cells decrease,
natural killer T cells (iNKT cells), which represent <1% of the T cell but it is unclear if their function is affected (191). As with T cells,
pool, decline in number but age-associated changes to their func- aging is associated with a decline in the numbers and proportions
tion have not been established (171). Monocyte numbers are stable of naïve B cells, an accumulation of memory B cells with limited
with aging, but classical cells (CD14++CD16−) decline, and inter- specificities, and impaired plasma cell antibody production (192).
mediate (CD14+CD16+) and non-classical (CD14+CD16++) cells Several robust and accepted hallmarks of immunosenescence
increase, but overall, monocyte cytokine production is impaired have been established, especially within the adaptive immune
(170, 172). These changes with blood monocytes are thought to system. For example, low numbers and proportions of naïve
be mirrored by tissue-resident macrophages, whereby classically T  cells (in particular CD8+ T  cells) and high numbers and
activated M1 cells decline, and alternatively activated M2 cells proportions of memory T cells (especially late-stage differenti-
accumulate (173). However, alterations in tissue-resident cells with ated CD8+ T  cells) are well established biomarkers (185, 186,
advancing age are very likely to be a result of adipose tissue accu- 193, 194). In addition, a cluster of parameters, revealed in
mulation and dysfunction that also occurs in parallel with aging longitudinal studies of octogenarians and nonagenarians from
(174, 175). Indeed, inflamed adipose tissue attracts macrophages an isolated population in Sweden, have been referred to as the
with cell-surface characteristics similar to M2 alternatively activated immune risk profile (195–197). Biomarkers included low num-
cells—often assumed to be anti-inflammatory. However, despite bers and proportions of B cells, high numbers and proportions
their cell-surface phenotype, these cells are potent producers of of late-stage differentiated CD8+ T  cells (i.e., CD27−CD28−),
inflammatory cytokines in adipose tissue, and likely drive age- and poor T  cell proliferation in response to mitogens, a CD4:CD8
obesity-associated inflammation (176–179). Thus, the M1/M2 ratio of <1.0, infection with Cytomegalovirus and high plasma
paradigm for macrophages is likely to be an over-simplification IL-6, which together, predicted greater all-cause mortality at
(180, 181). It is unknown if other, primarily tissue-resident cells, 2-, 4-, and 6-year follow-up (195, 197–200). Indeed, the age-
are affected by adipose tissue dysfunction, but with aging, the associated increase in systemic inflammation, referred to as
number and function of dendritic cells have been reported to “inflammageing” is another principle observation among aging
decrease in the skin and mucosal membranes (170). There is also and longevity studies (201, 202). Subsequently, high levels of
an age-associated increase in myeloid-derived suppressor cells—a IL-6, TNF-alpha, and C-reactive protein, have been associated
heterogeneous population of granulocytes, macrophages, and with shorter survival (203–205). Overall, it is well established
dendritic cells—that may impair aspects of immune function by that elderly individuals exhibit impaired immune responses to
producing reactive oxygen species and inhibitory cytokines (182). in vivo challenge with novel antigens (143, 206, 207) and these
Within the adaptive immune system, there are substantial individuals are subsequently thought to be at increased risk of
changes to the numbers, function, and phenotype of T cells with infection. Encouragingly however, as outlined in Part B: “Regular
aging. Among the broad population of CD4+ T-helper cells, aging Physical Activity and Frequent Exercise Augment Aspects of
is associated with a predominance of Th2 (i.e., IL-4 and IL-10), Immune Competency Across the Lifespan” exercise can be a
and Th17-producing cells (i.e., IL-17-producing cells that are potent stimulus of immune function, including the response
associated with autoimmune disease), whereas there is a decline to vaccination, and some evidence suggests that exercise might
in cells with a Th1 profile [i.e., IFN-gamma- and tumor necrosis delay or limit the age-associated decline in immune competency.
factor-alpha (TNF-alpha)-producing cells] (183, 184). With
aging, the numbers and proportions of antigen-inexperienced
CD4+ and CD8+ T cells decreases (e.g., CD27+CD28+CD45RA+
Relationships Between Exercise and
CD57−CD62L+CCR7+KLRG1− naïve cells) (185, 186). In parallel, Regular Physical Activity With Hallmarks
the numbers and proportions of antigen-experienced CD4+ and of an Aging Adaptive Immune System
CD8+ T  cells increases (e.g., CD27−CD28−CD45RA+CD57+CD As summarized in Part B: “Regular Physical Activity and Frequent
62L−CCR7−KLRG1+ memory cells), and these cells are potent Exercise Augment Aspects of Immune Competency Across the
producers of inflammatory cytokines (185, 186). These changes Lifespan” many studies have examined the influence of regular
are driven by lower hematopoietic stem cell numbers, thymic physical activity or frequent structured exercise on the function
involution resulting in reduced output of antigen-naïve T cells, of the adaptive immune system with aging (163–165). Here, we
and infection with latent viruses, in particular Cytomegalovirus focus on recent studies that have examined relationships between
(185, 187). With aging, T  cells that express natural killer cell- exercise, physical activity or cardiorespiratory fitness, and the
associated cell-surface proteins (NKT-like cells) also accumulate, numbers and proportions of CD4+ and CD8+ naïve and memory

Frontiers in Immunology  |  www.frontiersin.org 11 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

T  cells, as hallmarks of immunosenescence. Indeed, a small immune system. Most investigations have been cross-sectional
number of studies have investigated whether the characteristics in design, or have made observations between groups over
of the T cell pool are influenced by an active lifestyle. There is a short periods. For example, one study has shown that national
larger body of evidence in young adults, typically between 18 and standard triathletes (age range 18–36 years, n = 19), appear to
30 years, compared to older adults, hereafter considered as being exhibit impaired thymic output, assessed by T cell receptor exci-
over 40 years of age due to the characteristics of studies published to sion circle levels (211). Among CD4+ and CD8+ T cells, these
date. If an active lifestyle can be linked with a smaller accumulation athletes had lower absolute numbers of naïve cells and higher
of memory T cells, and a smaller decline in naïve T cells, then in absolute numbers of memory cells compared to age-matched
young adults, one interpretation might be that exercise prevents, or less active controls (n = 16), as shown by CD45RA, CD45RO,
at least delays immunosenescence, whereas in older adults, these and CD27 expression (211). Similar observations have been
associations could be interpreted as countering or limiting the made by examining individuals in the third decade of life, show-
development of an age-associated immune profile. ing that endurance athletes, compared to less active controls,
appear to exhibit slightly larger accumulations of memory
Experimental Evidence in Young People: Can T  cells and slightly fewer naïve T  cells, defined by CD45RA
Exercise Prevent or Delay Aging of the Adaptive and CCR7 expression (212). Thus, it appears that among
Immune System? younger individuals (i.e., less than 40 years of age), if exercise is
The characteristics of the T  cell pool have been examined in undertaken at very extreme volumes, such as more than 5–10
16 young adults (50% male; 18  ±  2  years) classified as being times the amount of physical activity recommended each week
very active (well-trained soccer players self-reporting 9–12  h (i.e., 12–25 h per week), this might contribute toward a small
of exercise per week) and compared to 16 young adults (50% decline in naïve T cells and a small increase in memory T cells.
male; 19  ±  2  years) classified as being untrained (individuals It is possible that these changes are due to reactivation of latent
self-reporting 2–3 h of exercise per week). Untrained individu- viruses, which could be independent of immune function, and
als showed the highest proportions of CD4+ and CD8+ memory driven by exercise-induced adrenergic activity, oxidative stress
T cells, and the lowest proportions of CD8+ naïve T cells, defined and inflammatory cytokines (48–50). However, these results
on the basis of CD57 and CD28 expression (208). Although these might also be explained by fluctuations in cell numbers and cell
results suggest that regular exercise might limit the age-associated sub-populations in peripheral blood over time. Such changes
accumulation of memory T cells and decline in naïve T cells, the have been interpreted as being linked to exercise training load
effects were strongly influenced by sex: only untrained males (213, 214), but it is also conceivable that these changes occur
exhibited high proportions of memory T cells and low propor- due to seasonal variation, as has been shown in non-exercise
tions of naïve cells compared to trained males, and these effects contexts (215, 216).
were driven by changes in the CD4+ T cell pool (208). Extending
these findings by examining a female-only population of young Experimental Evidence in Older People: Can
adults, the same authors compared 13 well-trained soccer players Exercise Limit or Counter Aging of the Adaptive
(self-reporting around 12 h of exercise per week; 20 ± 2 years) to Immune System?
13 untrained controls (self-reporting around 3 h of exercise per Although most studies have examined associations between
week; 21  ±  2  years) (209). Trained females exhibited a greater biomarkers of an aging adaptive immune system in young
proportion of CD8+ naïve T cells compared to untrained females, adults, other studies have made measurements across a broader
but these associations did not remain statistically significant after range of ages. For example, one study examined 102 men rang-
controlling for body fat percentage (trained 21.7  ±  4.0 versus ing in age from 18 to 61  years (mean 39  ±  6  years) (217). It
untrained 25.1 ± 4.1%, p < 0.05) (209). Indeed, very few studies was shown that the proportion of the CD4+ and CD8+ T  cell
have considered whether relationships between an active lifestyle pool comprising of memory cells (defined as KLRG1+CD57+
and markers of an aging immune system could be influenced by or KLRG1+CD28−) was inversely correlated with cardiores-
other factors, such as body composition. Recently, a very small piratory fitness, which is largely indicative of an active lifestyle
study of 15 males aged 30  ±  4  years, categorized participants (217). The age-associated decrease of naïve T  cells (defined as
using a combination of gold standard methods for measuring KLRG1−CD57− or KLRG1−CD28+) and increase in memory
physiological and lifestyle variables (210). Three groups were T cells did not withstand statistical adjustment for cardiorespira-
formed (sedentary, active, and very active) on the basis of tory fitness, but remained significant after adjusting statistically
objectively assessed habitual physical activity, directly measured for body composition and Cytomegalovirus infection (217).
cardiorespiratory fitness, and body composition assessed with Thus, it was concluded that fitter individuals exhibit a smaller
dual energy X-ray absorptiometry (210). This work showed that age-associated decline of naïve T cells and a smaller accumula-
sedentary individuals had higher proportions of memory CD4+ tion of memory T cells.
T cells expressing CD45RO and PD-1, supporting the results of As with work examining relationships between an active
other published studies that have not taken body composition lifestyle and hallmarks of an aging adaptive immune system in
into consideration. young and middle-aged adults, similar associations have been
It should be emphasized, that among younger adults in shown in an older population of 61 men aged 65–85  years
particular, mixed results have been reported when investigat- (218). In this study, participants self-reported to be “untrained”
ing links between an active lifestyle and hallmarks of an aging (n  =  15), or to lead a “moderate” training lifestyle (n  =  16;

Frontiers in Immunology  |  www.frontiersin.org 12 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

taking part in team sports or running less than 6 km two to from recent studies examining relationships between an active
three times per week), or an “intense” training lifestyle (n = 15; lifestyle and the characteristics of the T cell pool—as robust and
running approximately 10 km at least 5 days per week). These accepted biomarkers of immunosenescence—support obser-
categories were confirmed with a validated physical activity vations from some cross-sectional and longitudinal studies,
questionnaire and by measurement of cardiorespiratory fitness. showing that other measures of immune competency, which
Both training groups exhibited a lower proportion of CD4+ typically decline with aging, can be improved with physical
and CD8+ memory cells (defined as CD45RA+CCR7−), but activity or regular structured exercise (163–165). However,
these associations were largest and only statistically significant further research is needed in this area that employs precise
among men leading an “intense” training lifestyle, suggesting lifestyle measurements (e.g., using wearable technology to
a dose–response effect of exercise. Although findings were assess physical activity, and dual energy X-ray absorptiometry
less clear when examining other cell subpopulations based on to measure body composition) and more robust measurements
CD45RA and CCR7 expression, and there were no effects of of immune competency (e.g., absolute cell counts rather than
exercise when examining CD28− cells, men in the “trained” proportions, measurements of cell function, and in  vivo
groups had T cells with the longest telomeres (218). Another antigen challenges) while controlling for factors that drive
recent study compared 125 adults (55–79 years of age) who immunosenescence (e.g., inflammation and Cytomegalovirus
maintained a high level of cycling throughout life to 75 age- infection).
matched inactive controls (219). Within the CD4+ and CD8+
T cell pool, the frequency of naive cells (defined as CD45RA+) Mechanisms
was greater, and the frequency of memory cells (defined as Links between a physically active lifestyle with lower numbers
CD45RA-) was lower among cyclists. Extended phenotyping or proportions of memory T  cells, and higher numbers or
revealed that CD4+ and CD8+ CCR7-CD45RA+ accumulation proportions of naïve T cells, have been hypothesized as being
was less among cyclists, but no differences were found for driven by the acute effects of exercise bouts. For example, it has
CD28-CD57+ cells. Cyclists also exhibited higher frequencies been suggested that repeated bouts of exercise might prevent
of recent thymic emigrants and regulatory B cells, lower Th17 or delay immunological aging by limiting the accumulation of
polarization, and in plasma, higher IL-7 and lower IL-6 (219). CD4+ and CD8+ antigen-experienced memory T  cell clones,
Despite these findings, suggesting a beneficial effect of leading repopulating blood with antigen-inexperienced naïve T  cells
an active lifestyle on immunosenescence among older adults, (11, 12). In this hypothesis, it is proposed that memory T cells
there is some inconsistency in the literature. For example, are frequently mobilized into blood during regular bouts of
comparing elderly athletes (n = 12, approximately 74 years of exercise, followed by an extravasation to peripheral tissues,
age) to less active age-matched controls (n = 26), there were where these cells are exposed to pro-apoptotic stimuli, such
no differences in thymic output, the proportions of naïve as reactive oxygen species, glucocorticoids, and cytokines (11,
or memory CD4+ and CD8+ T  cells (defined with CD45RA 12). Subsequently, it is proposed that the number of naïve
and CCR7 expression), or T  cell activation in response to T cells increases as part of a negative feedback loop governing
anti-CD3 stimulation (212). Most investigations of T  cell the number of naïve and memory cells in the immune system,
immunosenescence and lifestyle among healthy elderly adults which is bolstered by exercise-induced thymopoiesis and extra-
have had cross-sectional study designs. Longitudinal studies, thymic T cell development (11, 12). Supporting the mechanisms
or randomized and controlled trials of exercise training are proposed in this hypothesis, many investigations have shown
lacking and might yield promising results. For example, one that memory T cells are mobilized into the circulation during
study has compared 6 months of exercise training in men and exercise, followed by extravasation out of the bloodstream
women (n  =  28, aged 61–76  years) to a similar group who in the hours following (81, 123). In addition, studies in mice
maintained their current lifestyle (n = 20, aged 62–79 years) show that lymphocyte apoptosis occurs post-exercise in tissues
(220). Using a simple immunophenotyping strategy, the results thought to be the homing destination of mobilized cells (222).
showed that the proportion of CD4+ T cells expressing CD28 Although some T cells mobilized by exercise might be resistant
increased in the exercise group after 6 months, but not in those to apoptosis, given that Cytomegalovirus-specific CD8+ T cells
who maintained their lifestyle (220). express high levels of Bcl-2 (223), other work has shown
that Cytomegalovirus-specific CD8+ T  cells, are equally as
Summary of Experimental Evidence susceptible to Fas-induced apoptosis as the total pool of CD8+
In summary, evidence shows that the characteristics of the T cells (224). Further, irrespective of virus specificity, studies
T cell pool appear to be influenced by leading an active life- have shown that T cells expressing cell-surface proteins such
style, determined by exercise training, physical activity level, as CD57 and KLRG1 are more susceptible to H2O2-induced
or cardiorespiratory fitness. It seems that among both the apoptosis than total lymphocytes and naïve T cells (225, 226).
young and elderly, an active lifestyle is generally linked to Thus, the concept of exercise directly countering memory
lower numbers and proportions of memory T cells and higher T cell accumulation is supported by evidence from human and
numbers and proportions of naïve T cells (10). This summary animal studies.
is partly supported by a recent systematic review, concluding It is unknown whether triggering apoptosis among expan­
that regular structured exercise increases the number of naïve ded clones of memory T  cells specific for viruses such as
T cells in peripheral blood at rest (221). Altogether, findings Cytomegalovirus is advantageous. For example, in a transplant

Frontiers in Immunology  |  www.frontiersin.org 13 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

setting, Cytomegalovirus disease occurs when T  cells fail to Thus, exercise might limit T cell immunosenescence by decreas-
provide antiviral control (227) and a robust pro-inflammatory ing visceral and subcutaneous adipose tissue (238), providing a
response to Cytomegalovirus has been associated with longer potent anti-inflammatory and anti-oxidative stimulus (247, 248).
survival in the elderly (228). However, it remains to be deter- In turn, lower systemic inflammation and better redox balance
mined what proportion of the T cell pool needs to be specific might limit viral reactivation, reducing stimulation with antigens
for Cytomegalovirus to limit viral reactivation. Infection with from viruses such as Cytomegalovirus. In addition, T cell dysfunc-
Cytomegalovirus results in approximately 10% of the CD4+ and tion might also be prevented, in part, by limiting reactive oxygen
CD8+ T cell pool becoming specific for this virus (229), although species production (249).
large inter-individual differences exist. For example, it has been In summary, leading a physically active lifestyle appears to
reported that 23% of the CD8+ T cell pool can become specific limit the age-associated changes to the cellular composition
for a single Cytomegalovirus epitope (223). Traditionally, it has of the adaptive immune system, but the mechanisms are yet
been considered disadvantageous for such a large proportion of to be determined. Exercise might counter the expansion of
the T cell pool to be specific for one virus. This view is linked memory T  cells directly, which is desirable assuming these
to another age- or infection-associated change that occurs in cells contribute to systemic inflammation and not all are
parallel—a fall in the numbers and proportions of naïve cells— required to control latent viruses. Limiting the expansion of
which has been interpreted as limiting capacity to engage novel memory T cells also assumes the “size” of the immune sys-
antigens. These interpretations are based upon two assumptions. tem is fixed, the capacity to produce antigen-naïve T cells is
First, there is an upper limit to the size of the immune system, limited, and these constraints contribute to immune decline
and second, thymic output is negligible after adolescence (230). in the elderly. However, exercise might affect memory T cell
Thus, it has been proposed that antigen-inexperienced naïve accumulation indirectly, by reducing viral reactivation, or
T cells could be “used up” due to ongoing differentiation into preventing T  cell senescence, by controlling adipose tissue
antigen-experienced memory T cells that “fill up” immunological deposition and dysfunction that drives inflammation and
“space” (230). It has also been proposed that this accumulation oxidative stress with aging and obesity.
of antigen-experienced memory T cells leads to “squeezing out”
of T cells targeting less dominant viruses leading to loss of viral CONCLUDING REMARKS
control (231). This concept of a fixed amount of immunological
space has since been debated (232, 233) and thymic output is Contemporary evidence from epidemiological studies shows
now known to persist, albeit reduced, up until around 70 years that leading a physically active lifestyle reduces the incidence
of age (234). However, even if removal of some memory T cells of communicable (e.g., bacterial and viral infections) and non-
is not essential for maintaining an effective T cell pool, assuming communicable diseases (e.g., cancer), implying that immune
these cells contribute to systemic inflammation, their removal competency is enhanced by regular exercise bouts. However, to
might limit “inflammageing” (230). Despite uncertainties over this day, research practice, academic teaching, and even physi-
the susceptibility of memory T  cells to undergo apoptosis, or cal activity promotion and prescription continues to consider a
whether it is advantageous to stimulate their removal, it seems prevailing myth that exercise can temporarily suppress immune
that exercise-induced immune cell death in the tissues has function. We have critically reviewed related evidence, and conclude
relevance to other processes. For example, apoptotic cells and that regular physical activity and frequent exercise are beneficial,
cell debris stimulates hematopoietic stem cell mobilization or at the very least, are not detrimental to immunological health.
into blood (95) perhaps promoting trafficking to the thymus We summarize that (i) limited reliable evidence exists to support
or extrathymic sites facilitating output of naïve T  cells (235). the claim that exercise suppresses cellular or soluble immune
Additional support for exercise stimulating production of naïve competency, (ii) exercise per se does not heighten the risk of
T  cells is provided by work showing that contracting skeletal opportunistic infections, and (iii) exercise can enhance in vivo
muscle produces IL-7 (236) which might increase thymic mass immune responses to bacterial, viral, and other antigens. In addi-
and function (237). tion, we present evidence showing that regular physical activity
A physically active lifestyle might also counter T cell immu- and frequent exercise might limit or delay immunological aging.
nosenescence indirectly, perhaps by limiting adipose tissue We conclude that leading an active lifestyle is likely to be benefi-
accumulation and dysfunction that occurs with aging and obesity cial, rather than detrimental, to immune function, which may
(174, 238, 239). Indeed, obesity has been linked with impaired have implications for health and disease in older age.
lymphocyte proliferation (240), shorter leukocyte telomere length
(241), and a skewing of the T cell pool toward a regulatory and AUTHOR CONTRIBUTIONS
Th2-phenotype (242). In addition, large expansions of differenti-
ated αβ T cells and γδ T cells have been shown among people with JC and JT contributed equally toward literature searching and
obesity, with γδ T  cells exhibiting impaired antiviral function retrieval, the ideas and interpretation of the studies described,
(243–245). It is generally accepted that repeated stimulation with drafting and revision of the manuscript, and approval of the final
antigens from Cytomegalovirus drives immunosenescence (185, version to be published. JT and JC both agreed to be accountable
186, 193, 194). With obesity, adipose tissue is the primary source for the work in ensuring that questions related to the accuracy or
of pro-inflammatory cytokines and reactive oxygen species (174, integrity of any part of the work are appropriately investigated
246) which can reactivate Cytomegalovirus directly (48, 49). and resolved.

Frontiers in Immunology  |  www.frontiersin.org 14 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

REFERENCES 22. Cox AJ, Gleeson M, Pyne DB, Callister R, Hopkins WG, Fricker PA. Clinical
and laboratory evaluation of upper respiratory symptoms in elite athletes.
1. Warburton DER, Bredin SSD. Health benefits of physical activity: a system- Clin J Sport Med (2008) 18(5):438–45. doi:10.1097/JSM.0b013e318181e501
atic review of current systematic reviews. Curr Opin Cardiol (2017) 32(5): 23. Makela MJ, Puhakka T, Ruuskanen O, Leinonen M, Saikku P, Kimpimaki M,
541–56. doi:10.1097/HCO.0000000000000437 et al. Viruses and bacteria in the etiology of the common cold. J Clin Microbiol
2. Pape K, Ryttergaard L, Rotevatn TA, Nielsen BJ, Torp-Pedersen C, (1998) 36(2):539–42.
Overgaard C, et al. Leisure-time physical activity and the risk of suspected 24. Nieman DC. Exercise, infection, and immunity. Int J Sports Med (1994)
bacterial infections. Med Sci Sports Exerc (2016) 48(9):1737–44. doi:10.1249/ 15(Suppl 3):S131–41. doi:10.1055/s-2007-1021128
MSS.0000000000000953 25. Meeusen R, Duclos M, Foster C, Fry A, Gleeson M, Nieman D, et  al.
3. Kostka T, Berthouze SE, Lacour J, Bonnefoy M. The symptomatology of Prevention, diagnosis and treatment of the overtraining syndrome: joint
upper respiratory tract infections and exercise in elderly people. Med Sci consensus statement of the European College of Sport Science (ECSS) and
Sports Exerc (2000) 32(1):46–51. doi:10.1097/00005768-200001000-00008 the American College of Sports Medicine (ACSM). Eur J Sport Sci (2013)
4. Leveille SG, Gray S, LaCroix AZ, Ferrucci L, Black DJ, Guralnik JM. Physical 13(1):1–24. doi:10.1080/17461391.2012.730061
inactivity and smoking increase risk for serious infections in older women. 26. Meeusen R, Duclos M, Foster C, Fry A, Gleeson M, Nieman D, et  al.
J Am Geriatr Soc (2000) 48(12):1582–8. doi:10.1111/j.1532-5415.2000. Prevention, diagnosis, and treatment of the overtraining syndrome:
tb03867.x joint consensus statement of the European College of Sport Science and
5. Romaniszyn D, Pobiega M, Wojkowska-Mach J, Chmielarczyk A, Gryglewska B, the American College of Sports Medicine. Med Sci Sports Exerc (2013)
Adamski P, et al. The general status of patients and limited physical activity 45(1):186–205. doi:10.1249/MSS.0b013e318279a10a
as risk factors of methicillin-resistant Staphylococcus aureus occurrence 27. Edwards JP, Walsh NP, Diment PC, Roberts R. Anxiety and perceived
in long-term care facilities residents in Krakow, Poland. BMC Infect Dis psychological stress play an important role in the immune response after
(2014) 14:271. doi:10.1186/1471-2334-14-271 exercise. Exerc Immunol Rev (2018) 24:26–34.
6. Baik I, Curhan GC, Rimm EB, Bendich A, Willett WC, Fawzi WW. 28. Glaser R, Kiecolt-Glaser JK. Stress-induced immune dysfunction: implica-
A prospective study of age and lifestyle factors in relation to community- tions for health. Nat Rev Immunol (2005) 5(3):243–51. doi:10.1038/nri1571
acquired pneumonia in US men and women. Arch Intern Med (2000) 29. Kohut ML, Lee W, Martin A, Arnston B, Russell DW, Ekkekakis P, et al. The
160(20):3082–8. doi:10.1001/archinte.160.20.3082 exercise-induced enhancement of influenza immunity is mediated in part by
7. Walsh NP, Gleeson M, Pyne DB, Nieman DC, Dhabhar FS, Shephard RJ, et al. improvements in psychosocial factors in older adults. Brain Behav Immun
Position statement. Part two: maintaining immune health. Exerc Immunol (2005) 19(4):357–66. doi:10.1016/j.bbi.2004.12.002
Rev (2011) 17:64–103. 30. Bermon S, Castell LM, Calder PC, Bishop NC, Blomstrand E, Mooren FC,
8. Walsh NP, Gleeson M, Shephard RJ, Gleeson M, Woods JA, Bishop NC, et  al. Consensus statement immunonutrition and exercise. Exerc Immunol
et  al. Position statement. Part one: immune function and exercise. Exerc Rev (2017) 23:8–50.
Immunol Rev (2011) 17:6–63. 31. Choudhry AJ, Al-Mudaimegh KS, Turkistani AM, Al-Hamdan NA. Hajj-
9. Pascoe AR, Fiatarone Singh MA, Edwards KM. The effects of exercise associated acute respiratory infection among hajjis from Riyadh. East
on vaccination responses: a review of chronic and acute exercise inter- Mediterr Health J (2006) 12(3–4):300–9.
ventions in humans. Brain Behav Immun (2014) 39:33–41. doi:10.1016/j. 32. Svendsen IS, Taylor IM, Tonnessen E, Bahr R, Gleeson M. Training-related
bbi.2013.10.003 and competition-related risk factors for respiratory tract and gastroin-
10. Turner JE. Is immunosenescence influenced by our lifetime “dose” of testinal infections in elite cross-country skiers. Br J Sports Med (2016)
exercise? Biogerontology (2016) 17(3):581–602. doi:10.1007/s10522-016- 50(13):809–15. doi:10.1136/bjsports-2015-095398
9642-z 33. Gleeson M. Immunological aspects of sport nutrition. Immunol Cell Biol
11. Simpson RJ, Guy K. Coupling aging immunity with a sedentary lifestyle: (2016) 94(2):117–23. doi:10.1038/icb.2015.109
has the damage already been done? – a mini-review. Gerontology (2010) 34. Schwellnus MP, Derman WE, Jordaan E, Page T, Lambert MI, Readhead C,
56(5):449–58. doi:10.1159/000270905 et  al. Elite athletes travelling to international destinations >5 time zone
12. Simpson RJ. Aging, persistent viral infections, and immunosenescence: can differences from their home country have a 2-3-fold increased risk of illness.
exercise “make space”? Exerc Sport Sci Rev (2011) 39(1):23–33. doi:10.1097/ Br J Sports Med (2012) 46(11):816–21. doi:10.1136/bjsports-2012-091395
JES.0b013e318201f39d 35. Fondell E, Lagerros YT, Sundberg CJ, Lekander M, Balter O, Rothman KJ,
13. Turner JE, Brum PC. Does regular exercise counter T  cell immunosenes- et  al. Physical activity, stress, and self-reported upper respiratory tract
cence reducing the risk of developing cancer and promoting successful infection. Med Sci Sports Exerc (2011) 43(2):272–9. doi:10.1249/MSS.
treatment of malignancies? Oxid Med Cell Longev (2017) 2017:4234765. 0b013e3181edf108
doi:10.1155/2017/4234765 36. Martensson S, Nordebo K, Malm C. High training volumes are associated
14. Mackinnon LT. Advances in Exercise Immunology. Champaign, IL: Human with a low number of self-reported sick days in elite endurance athletes.
Kinetics (1999). xii, 363 p. J Sports Sci Med (2014) 13(4):929–33.
15. Peake JM, Neubauer O, Walsh NP, Simpson RJ. Recovery of the immune sys- 37. Hellard P, Avalos M, Guimaraes F, Toussaint JF, Pyne DB. Training-related
tem after exercise. J Appl Physiol (1985) (2017) 122(5):1077–87. doi:10.1152/ risk of common illnesses in elite swimmers over a 4-yr period. Med Sci Sports
japplphysiol.00622.2016 Exerc (2015) 47(4):698–707. doi:10.1249/MSS.0000000000000461
16. Cowles WN. Fatigue as a contributory cause of pneumonias. Boston Med 38. Hoffman MD, Krishnan E. Health and exercise-related medical issues among
Surg J (1918) 179:555. doi:10.1056/NEJM191810311791801 1,212 ultramarathon runners: baseline findings from the Ultrarunners
17. Peters EM, Bateman ED. Ultramarathon running and upper respiratory tract Longitudinal TRAcking (ULTRA) study. PLoS One (2014) 9(1):e83867.
infections. An epidemiological survey. S Afr Med J (1983) 64(15):582–4. doi:10.1371/journal.pone.0083867
18. Nieman DC, Johanssen LM, Lee JW, Arabatzis K. Infectious episodes in 39. Hoffman MD, Fogard K. Demographic characteristics of 161-km ultramar-
runners before and after the Los Angeles Marathon. J Sports Med Phys Fitness athon runners. Res Sports Med (2012) 20(1):59–69. doi:10.1080/15438627.2
(1990) 30(3):316–28. 012.634707
19. Nieman DC, Johanssen LM, Lee JW. Infectious episodes in runners before 40. Malm C. Susceptibility to infections in elite athletes: the S-curve. Scand
and after a roadrace. J Sports Med Phys Fitness (1989) 29(3):289–96. J Med Sci Sports (2006) 16(1):4–6. doi:10.1111/j.1600-0838.2005.00499.x
20. Spence L, Brown WJ, Pyne DB, Nissen MD, Sloots TP, McCormack JG, 41. Derman W, Schwellnus M, Jordaan E. Clinical characteristics of 385 illnesses
et al. Incidence, etiology, and symptomatology of upper respiratory illness of athletes with impairment reported on the WEB-IISS system during
in elite athletes. Med Sci Sports Exerc (2007) 39(4):577–86. doi:10.1249/ the London 2012 Paralympic Games. PM R (2014) 6(8 Suppl):S23–30.
mss.0b013e31802e851a doi:10.1016/j.pmrj.2014.05.016
21. Gleeson M. Immune function in sport and exercise. J Appl Physiol (1985) 42. Bonini M, Gramiccioni C, Fioretti D, Ruckert B, Rinaldi M, Akdis C,
(2007) 103(2):693–9. doi:10.1152/japplphysiol.00008.2007 et al. Asthma, allergy and the Olympics: a 12-year survey in elite athletes.

Frontiers in Immunology  |  www.frontiersin.org 15 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

Curr Opin Allergy Clin Immunol (2015) 15(2):184–92. doi:10.1097/ACI. 62. Allgrove JE, Gomes E, Hough J, Gleeson M. Effects of exercise intensity on
0000000000000149 salivary antimicrobial proteins and markers of stress in active men. J Sports
43. Bramley TJ, Lerner D, Sames M. Productivity losses related to the common Sci (2008) 26(6):653–61. doi:10.1080/02640410701716790
cold. J Occup Environ Med (2002) 44(9):822–9. doi:10.1097/00043764- 63. Sari-Sarraf V, Reilly T, Doran DA. Salivary IgA response to intermittent and
200209000-00004 continuous exercise. Int J Sports Med (2006) 27(11):849–55. doi:10.1055/
44. Fendrick AM, Monto AS, Nightengale B, Sarnes M. The economic burden of s-2006-923777
non-influenza-related viral respiratory tract infection in the United States. 64. Reid MR, Drummond PD, Mackinnon LT. The effect of moderate aerobic
Arch Intern Med (2003) 163(4):487–94. doi:10.1001/archinte.163.4.487 exercise and relaxation on secretory immunoglobulin A. Int J Sports Med
45. Bayer C, Remschmidt C, an der Heiden M, Tolksdorf K, Herzhoff M, (2001) 22(2):132–7. doi:10.1055/s-2001-11347
Kaersten S, et al. Internet-based syndromic monitoring of acute respiratory 65. Davison G. Innate immune responses to a single session of sprint interval
illness in the general population of Germany, weeks 35/2011 to 34/2012. Euro training. Appl Physiol Nutr Metab (2011) 36(3):395–404. doi:10.1139/h11-033
Surveill (2014) 19(4):1–11. doi:10.2807/1560-7917.ES2014.19.4.20684 66. Walsh NP, Blannin AK, Clark AM, Cook L, Robson PJ, Gleeson M. The effects
46. Gleeson M, Pyne DB, Austin JP, Lynn Francis J, Clancy RL, McDonald WA, of high-intensity intermittent exercise on saliva IgA, total protein and alpha-
et  al. Epstein-Barr virus reactivation and upper-respiratory illness in amylase. J Sports Sci (1999) 17(2):129–34. doi:10.1080/026404199366226
elite swimmers. Med Sci Sports Exerc (2002) 34(3):411–7. doi:10.1097/ 67. Peters EM, Shaik J, Kleinveldt N. Upper respiratory tract infection symp-
00005768-200203000-00005 toms in ultramarathon runners not related to immunoglobulin status. Clin
47. Cox AJ, Gleeson M, Pyne DB, Saunders PU, Clancy RL, Fricker PA. Valtrex J Sport Med (2010) 20(1):39–46. doi:10.1097/JSM.0b013e3181cb4086
therapy for Epstein-Barr virus reactivation and upper respiratory symptoms 68. Gleeson M, Bishop N, Oliveira M, McCauley T, Tauler P, Muhamad AS.
in elite runners. Med Sci Sports Exerc (2004) 36(7):1104–10. doi:10.1249/01. Respiratory infection risk in athletes: association with antigen-stimulated
MSS.0000131957.40985.2B IL-10 production and salivary IgA secretion. Scand J Med Sci Sports (2012)
48. Speir E. Cytomegalovirus gene regulation by reactive oxygen species. Agents 22(3):410–7. doi:10.1111/j.1600-0838.2010.01272.x
in atherosclerosis. Ann N Y Acad Sci (2000) 899:363–74. doi:10.1111/j. 69. Gleeson M, Bishop N, Oliveira M, Tauler P. Influence of training load on
1749-6632.2000.tb06200.x upper respiratory tract infection incidence and antigen-stimulated cytokine
49. Docke WD, Prosch S, Fietze E, Kimel V, Zuckermann H, Klug C, et  al. production. Scand J Med Sci Sports (2013) 23(4):451–7. doi:10.1111/j.
Cytomegalovirus reactivation and tumour necrosis factor. Lancet (1994) 1600-0838.2011.01422.x
343(8892):268–9. doi:10.1016/S0140-6736(94)91116-9 70. Brandtzaeg P. Secretory immunity with special reference to the oral cavity.
50. Prösch S, Wendt CEC, Reinke P, Priemer C, Oppert M, Krüger DH, et al. J Oral Microbiol (2013) 5:1–24. doi:10.3402/jom.v5i0.20401
A novel link between stress and human cytomegalovirus (HCMV) infection: 71. Marcotte H, Lavoie MC. Oral microbial ecology and the role of salivary
sympathetic hyperactivity stimulates HCMV activation. Virology (2000) immunoglobulin A. Microbiol Mol Biol Rev (1998) 62(1):71–109.
272:357–65. doi:10.1006/viro.2000.0367 72. Needleman I, Ashley P, Petrie A, Fortune F, Turner W, Jones J, et  al. Oral
51. He CS, Handzlik M, Muhamad A, Gleeson M. Influence of CMV/EBV health and impact on performance of athletes participating in the London
serostatus on respiratory infection incidence during 4 months of winter 2012 Olympic Games: a cross-sectional study. Br J Sports Med (2013)
training in a student cohort of endurance athletes. Eur J Appl Physiol (2013) 47(16):1054–8. doi:10.1136/bjsports-2013-092891
113(10):2613–9. doi:10.1007/s00421-013-2704-x 73. Heaney JL, Gleeson M, Phillips AC, Taylor IM, Drayson MT, Goodall M,
52. Bishop NC, Gleeson M. Acute and chronic effects of exercise on markers et  al. Salivary immunoglobulin free light chains: reference ranges and
of mucosal immunity. Front Biosci (Landmark Ed) (2009) 14:4444–56. responses to exercise in young and older adults. Exerc Immunol Rev
doi:10.2741/3540 (2016) 22:28–41.
53. Tomasi TB, Trudeau FB, Czerwinski D, Erredge S. Immune parameters 74. Li TL, Gleeson M. The effect of single and repeated bouts of prolonged
in athletes before and after strenuous exercise. J Clin Immunol (1982) cycling and circadian variation on saliva flow rate, immunoglobulin A and
2(3):173–8. doi:10.1007/BF00915219 alpha-amylase responses. J Sports Sci (2004) 22(11–12):1015–24. doi:10.108
54. Mackinnon LT, Chick TW, van As A, Tomasi TB. Decreased secretory 0/02640410410001716733
immunoglobulins following intense endurance exercise. Sports Train Med 75. Engeland CG, Hugo FN, Hilgert JB, Nascimento GG, Junges R, Lim HJ,
Rehabil (1989) 1:209–18. doi:10.1080/15438628909511878 et  al. Psychological distress and salivary secretory immunity. Brain Behav
55. Nieman DC, Henson DA, Fagoaga OR, Utter AC, Vinci DM, Davis JM, et al. Immun (2016) 52:11–7. doi:10.1016/j.bbi.2015.08.017
Change in salivary IgA following a competitive marathon race. Int J Sports 76. Deslauriers N, Oudghiri M, Seguin J, Trudel L. The oral immune system:
Med (2002) 23(1):69–75. doi:10.1055/s-2002-19375 dynamics of salivary immunoglobulin production in the inbred mouse.
56. Nehlsen-Cannarella SL, Nieman DC, Fagoaga OR, Kelln WJ, Henson DA, Immunol Invest (1986) 15(4):339–49. doi:10.3109/08820138609052953
Shannon M, et al. Saliva immunoglobulins in elite women rowers. Eur J Appl 77. Booth CK, Dwyer DB, Pacque PF, Ball MJ. Measurement of immunoglob-
Physiol (2000) 81(3):222–8. doi:10.1007/s004210050034 ulin A in saliva by particle-enhanced nephelometric immunoassay: sample
57. Palmer FM, Nieman DC, Henson DA, McAnulty SR, McAnulty L, Swick NS, collection, limits of quantitation, precision, stability and reference range.
et al. Influence of vitamin C supplementation on oxidative and salivary IgA Ann Clin Biochem (2009) 46(Pt 5):401–6. doi:10.1258/acb.2009.008248
changes following an ultramarathon. Eur J Appl Physiol (2003) 89(1):100–7. 78. Brandtzaeg P. Do salivary antibodies reliably reflect both mucosal and
doi:10.1007/s00421-002-0756-4 systemic immunity? Ann N Y Acad Sci (2007) 1098:288–311. doi:10.1196/
58. Steerenberg PA, van Asperen IA, van Nieuw Amerongen A, Biewenga A, annals.1384.012
Mol D, Medema GJ. Salivary levels of immunoglobulin A in triathletes. 79. Hanstock HG, Walsh NP, Edwards JP, Fortes MB, Cosby SL, Nugent A,
Eur J  Oral Sci (1997) 105(4):305–9. doi:10.1111/j.1600-0722.1997. et al. Tear fluid SIgA as a noninvasive biomarker of mucosal immunity and
tb00245.x common cold risk. Med Sci Sports Exerc (2016) 48(3):569–77. doi:10.1249/
59. Nieman DC, Dumke CI, Henson DA, McAnulty SR, McAnulty LS, MSS.0000000000000801
Lind RH, et  al. Immune and oxidative changes during and following 80. Gleeson M, Pyne DB, Elkington LJ, Hall ST, Attia JR, Oldmeadow C, et al.
the western states endurance run. Int J Sports Med (2003) 24(7):541–7. Developing a multi-component immune model for evaluating the risk of
doi:10.1055/s-2003-42018 respiratory illness in athletes. Exerc Immunol Rev (2017) 23:52–64.
60. Blannin AK, Robson PJ, Walsh NP, Clark AM, Glennon L, Gleeson M. The 81. Campbell JP, Riddell NE, Burns VE, Turner M, van Zanten JJ, Drayson MT,
effect of exercising to exhaustion at different intensities on saliva immu- et al. Acute exercise mobilises CD8+ T lymphocytes exhibiting an effector-
noglobulin A, protein and electrolyte secretion. Int J Sports Med (1998) memory phenotype. Brain Behav Immun (2009) 23(6):767–75. doi:10.1016/j.
19(8):547–52. doi:10.1055/s-2007-971958 bbi.2009.02.011
61. Killer SC, Svendsen IS, Gleeson M. The influence of hydration status 82. Shephard RJ. Adhesion molecules, catecholamines and leucocyte redis-
during prolonged endurance exercise on salivary antimicrobial proteins. tribution during and following exercise. Sports Med (2003) 33(4):261–84.
Eur J Appl Physiol (2015) 115(9):1887–95. doi:10.1007/s00421-015-3173-1 doi:10.2165/00007256-200333040-00002

Frontiers in Immunology  |  www.frontiersin.org 16 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

83. Benschop RJ, Nijkamp FP, Ballieux RE, Heijnen CJ. The effects of beta- 104. Poli A, Michel T, Theresine M, Andres E, Hentges F, Zimmer J. CD56bright
adrenoceptor stimulation on adhesion of human natural killer cells to natural killer (NK) cells: an important NK cell subset. Immunology (2009)
cultured endothelium. Br J Pharmacol (1994) 113(4):1311–6. doi:10.1111/j. 126(4):458–65. doi:10.1111/j.1365-2567.2008.03027.x
1476-5381.1994.tb17141.x 105. Bjorkstrom NK, Riese P, Heuts F, Andersson S, Fauriat C, Ivarsson MA, et al.
84. Kruger K, Lechtermann A, Fobker M, Volker K, Mooren FC. Exercise-induced Expression patterns of NKG2A, KIR, and CD57 define a process of CD56dim
redistribution of T  lymphocytes is regulated by adrenergic mechanisms. NK-cell differentiation uncoupled from NK-cell education. Blood (2010)
Brain Behav Immun (2008) 22(3):324–38. doi:10.1016/j.bbi.2007.08.008 116(19):3853–64. doi:10.1182/blood-2010-04-281675
85. Dimitrov S, Lange T, Born J. Selective mobilization of cytotoxic leukocytes 106. Lopez-Verges S, Milush JM, Pandey S, York VA, Arakawa-Hoyt J, Pircher H,
by epinephrine. J Immunol (2010) 184(1):503–11. doi:10.4049/jimmunol. et al. CD57 defines a functionally distinct population of mature NK cells in
0902189 the human CD56dimCD16+ NK-cell subset. Blood (2010) 116(19):3865–74.
86. Turner JE, Spielmann G, Wadley AJ, Aldred S, Simpson RJ, Campbell JP. doi:10.1182/blood-2010-04-282301
Exercise-induced B cell mobilisation: preliminary evidence for an influx of 107. Bigley AB, Rezvani K, Chew C, Sekine T, Pistillo M, Crucian B, et al. Acute
immature cells into the bloodstream. Physiol Behav (2016) 164(Pt A):376–82. exercise preferentially redeploys NK-cells with a highly-differentiated
doi:10.1016/j.physbeh.2016.06.023 phenotype and augments cytotoxicity against lymphoma and multiple
87. Kruger K, Alack K, Ringseis R, Mink L, Pfeifer E, Schinle M, et al. Apoptosis myeloma target cells. Brain Behav Immun (2014) 39:160–71. doi:10.1016/j.
of T-cell subsets after acute high-intensity interval exercise. Med Sci Sports bbi.2013.10.030
Exerc (2016) 48(10):2021–9. doi:10.1249/MSS.0000000000000979 108. Bigley AB, Rezvani K, Pistillo M, Reed J, Agha N, Kunz H, et al. Acute exercise
88. Clifford T, Wood MJ, Stocks P, Howatson G, Stevenson EJ, Hilkens CMU. preferentially redeploys NK-cells with a highly-differentiated phenotype and
T-regulatory cells exhibit a biphasic response to prolonged endurance augments cytotoxicity against lymphoma and multiple myeloma target cells.
exercise in humans. Eur J Appl Physiol (2017) 117(8):1727–37. doi:10.1007/ Part II: impact of latent cytomegalovirus infection and catecholamine sen-
s00421-017-3667-0 sitivity. Brain Behav Immun (2015) 49:59–65. doi:10.1016/j.bbi.2014.12.027
89. Shek PN, Sabiston BH, Buguet A, Radomski MW. Strenuous exercise and 109. Simpson RJ, Florida-James GD, Cosgrove C, Whyte GP, Macrae S,
immunological changes: a multiple-time-point analysis of leukocyte subsets, Pircher H, et al. High-intensity exercise elicits the mobilization of senescent
CD4/CD8 ratio, immunoglobulin production and NK  cell response. Int T lymphocytes into the peripheral blood compartment in human subjects.
J Sports Med (1995) 16(7):466–74. doi:10.1055/s-2007-973039 J Appl Physiol (1985) (2007) 103(1):396–401. doi:10.1152/japplphysiol.
90. Shinkai S, Shore S, Shek PN, Shephard RJ. Acute exercise and immune 00007.2007
function. Relationship between lymphocyte activity and changes 110. Simpson RJ, Cosgrove C, Ingram LA, Florida-James GD, Whyte GP, Pircher H,
in subset counts. Int J Sports Med (1992) 13(6):452–61. doi:10.105 et  al. Senescent T-lymphocytes are mobilised into the peripheral blood
5/s-2007-1021297 compartment in young and older humans after exhaustive exercise. Brain
91. Kakanis MW, Peake J, Brenu EW, Simmonds M, Gray B, Hooper SL, et al. Behav Immun (2008) 22(4):544–51. doi:10.1016/j.bbi.2007.11.002
The open window of susceptibility to infection after acute exercise in healthy 111. Simpson RJ, Cosgrove C, Chee MM, McFarlin BK, Bartlett DB, Spielmann G,
young male elite athletes. Exerc Immunol Rev (2010) 16:119–37. et  al. Senescent phenotypes and telomere lengths of peripheral blood
92. Pedersen BK, Ullum H. NK  cell response to physical activity: possible T-cells mobilized by acute exercise in humans. Exerc Immunol Rev (2010)
mechanisms of action. Med Sci Sports Exerc (1994) 26(2):140–6. doi:10.1249/ 16:40–55.
00005768-199402000-00003 112. Pedersen L, Idorn M, Olofsson GH, Lauenborg B, Nookaew I, Hansen RH,
93. Dhabhar FS. Effects of stress on immune function: the good, the bad, and et al. Voluntary running suppresses tumor growth through epinephrine- and
the beautiful. Immunol Res (2014) 58(2–3):193–210. doi:10.1007/s12026- IL-6-dependent NK cell mobilization and redistribution. Cell Metab (2016)
014-8517-0 23(3):554–62. doi:10.1016/j.cmet.2016.01.011
94. Kruger K, Mooren FC. T cell homing and exercise. Exerc Immunol Rev (2007) 113. Kim R, Emi M, Tanabe K. Cancer immunoediting from immune surveillance
13:37–54. to immune escape. Immunology (2007) 121(1):1–14. doi:10.1111/j.1365-
95. Mooren FC, Kruger K. Apoptotic lymphocytes induce progenitor cell 2567.2007.02587.x
mobilization after exercise. J Appl Physiol (1985) (2015) 119(2):135–9. 114. Scheiermann C, Kunisaki Y, Frenette PS. Circadian control of the immune
doi:10.1152/japplphysiol.00287.2015 system. Nat Rev Immunol (2013) 13(3):190–8. doi:10.1038/nri3386
96. Mars M, Govender S, Weston A, Naicker V, Chuturgoon A. High intensity 115. Suzuki K, Hayano Y, Nakai A, Furuta F, Noda M. Adrenergic control of the
exercise: a cause of lymphocyte apoptosis? Biochem Biophys Res Commun adaptive immune response by diurnal lymphocyte recirculation through
(1998) 249(2):366–70. doi:10.1006/bbrc.1998.9156 lymph nodes. J Exp Med (2016) 213(12):2567–74. doi:10.1084/jem.20160723
97. Mooren FC, Lechtermann A, Volker K. Exercise-induced apoptosis of 116. Dimitrov S, Benedict C, Heutling D, Westermann J, Born J, Lange T. Cortisol
lymphocytes depends on training status. Med Sci Sports Exerc (2004) and epinephrine control opposing circadian rhythms in T cell subsets. Blood
36(9):1476–83. doi:10.1249/01.MSS.0000139897.34521.E9 (2009) 113(21):5134–43. doi:10.1182/blood-2008-11-190769
98. Mooren FC, Bloming D, Lechtermann A, Lerch MM, Volker K. Lymphocyte 117. Hojman P. Exercise protects from cancer through regulation of immune
apoptosis after exhaustive and moderate exercise. J Appl Physiol (2002) function and inflammation. Biochem Soc Trans (2017) 45(4):905–11.
93(1):147–53. doi:10.1152/japplphysiol.01262.2001 doi:10.1042/BST20160466
99. Tanimura Y, Shimizu K, Tanabe K, Otsuki T, Yamauchi R, Matsubara Y, et al. 118. Turner JE, Wadley AJ, Aldred S, Fisher JP, Bosch JA, Campbell JP. Inten­
Exercise-induced oxidative DNA damage and lymphocytopenia in seden- sive exercise does not preferentially mobilize skin-homing T  cells and
tary young males. Med Sci Sports Exerc (2008) 40(8):1455–62. doi:10.1249/ NK  cells. Med Sci Sports Exerc (2016) 48(7):1285–93. doi:10.1249/MSS.
MSS.0b013e31817242cf 0000000000000914
100. Simpson RJ, Florida-James GD, Whyte GP, Black JR, Ross JA, Guy K. 119. Hanson ED, Danson E, Nguyen-Robertson CV, Fyfe JJ, Stepto NK,
Apoptosis does not contribute to the blood lymphocytopenia observed Bartlett DB, et al. Maximal exercise increases mucosal associated invariant
after intensive and downhill treadmill running in humans. Res Sports Med T  cell frequency and number in healthy young men. Eur J Appl Physiol
(2007) 15(3):157–74. doi:10.1080/15438620701405339 (2017) 117(11):2159–69. doi:10.1007/s00421-017-3704-z
101. Shephard RJ, Shek PN. Effects of exercise and training on natural killer 120. Gross E, Sunwoo JB, Bui JD. Cancer immunosurveillance and immu-
cell counts and cytolytic activity: a meta-analysis. Sports Med (1999) 28(3): noediting by natural killer cells. Cancer J (2013) 19(6):483–9. doi:10.1097/
177–95. doi:10.2165/00007256-199928030-00003 PPO.0000000000000005
102. Hansen JB, Wilsgard L, Osterud B. Biphasic changes in leukocytes induced 121. Simpson RJ, Bigley AB, Agha N, Hanley PJ, Bollard CM. Mobilizing immune
by strenuous exercise. Eur J Appl Physiol Occup Physiol (1991) 62(3):157–61. cells with exercise for cancer immunotherapy. Exerc Sport Sci Rev (2017)
doi:10.1007/BF00643735 45(3):163–72. doi:10.1249/JES.0000000000000114
103. Freud AG, Caligiuri MA. Human natural killer cell development. Immunol 122. Moore SC, Lee IM, Weiderpass E, Campbell PT, Sampson JN, Kitahara CM,
Rev (2006) 214:56–72. doi:10.1111/j.1600-065X.2006.00451.x et  al. Association of leisure-time physical activity with risk of 26 types of

Frontiers in Immunology  |  www.frontiersin.org 17 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

cancer in 1.44 million adults. JAMA Intern Med (2016) 176(6):816–25. of influenza vaccination in older adults. Brain Behav Immun (2015)
doi:10.1001/jamainternmed.2016.1548 49:e24–5. doi:10.1016/j.bbi.2015.06.102
123. Turner JE, Aldred S, Witard OC, Drayson MT, Moss PM, Bosch JA. Latent 141. Furman D, Hejblum BP, Simon N, Jojic V, Dekker CL, Thiebaut R, et  al.
cytomegalovirus infection amplifies CD8 T-lymphocyte mobilisation and Systems analysis of sex differences reveals an immunosuppressive role for
egress in response to exercise. Brain Behav Immun (2010) 24(8):1362–70. testosterone in the response to influenza vaccination. Proc Natl Acad Sci
doi:10.1016/j.bbi.2010.07.239 U S A (2014) 111(2):869–74. doi:10.1073/pnas.1321060111
124. Lancaster GI, Khan Q, Drysdale PT, Wallace F, Jeukendrup AE, Drayson MT, 142. Trigunaite A, Dimo J, Jorgensen TN. Suppressive effects of androgens on
et  al. Effect of prolonged exercise and carbohydrate ingestion on type 1 the immune system. Cell Immunol (2015) 294(2):87–94. doi:10.1016/j.
and type 2 T  lymphocyte distribution and intracellular cytokine produc- cellimm.2015.02.004
tion in humans. J Appl Physiol (1985) (2005) 98(2):565–71. doi:10.1152/ 143. Haq K, McElhaney JE. Immunosenescence: influenza vaccination and
japplphysiol.00754.2004 the elderly. Curr Opin Immunol (2014) 29:38–42. doi:10.1016/j.coi.2014.
125. Starkie RL, Rolland J, Febbraio MA. Effect of adrenergic blockade on lym- 03.008
phocyte cytokine production at rest and during exercise. Am J Physiol Cell 144. Kohut ML, Cooper MM, Nickolaus MS, Russell DR, Cunnick JE. Exercise
Physiol (2001) 281(4):C1233–40. doi:10.1152/ajpcell.2001.281.4.C1233 and psychosocial factors modulate immunity to influenza vaccine in elderly
126. Siedlik JA, Deckert JA, Benedict SH, Bhatta A, Dunbar AJ, Vardiman JP, individuals. J Gerontol A Biol Sci Med Sci (2002) 57(9):M557–62. doi:10.1093/
et al. T cell activation and proliferation following acute exercise in human gerona/57.9.M557
subjects is altered by storage conditions and mitogen selection. J Immunol 145. Kohut ML, Arntson BA, Lee W, Rozeboom K, Yoon KJ, Cunnick JE, et al.
Methods (2017) 446:7–14. doi:10.1016/j.jim.2017.03.017 Moderate exercise improves antibody response to influenza immuniza-
127. Siedlik JA, Benedict SH, Landes EJ, Weir JP, Vardiman JP, Gallagher PM. tion in older adults. Vaccine (2004) 22(17–18):2298–306. doi:10.1016/j.
Acute bouts of exercise induce a suppressive effect on lymphocyte prolif- vaccine.2003.11.023
eration in human subjects: a meta-analysis. Brain Behav Immun (2016) 146. Smith TP, Kennedy SL, Fleshner M. Influence of age and physical activity on
56:343–51. doi:10.1016/j.bbi.2016.04.008 the primary in vivo antibody and T cell-mediated responses in men. J Appl
128. Zimmer P, Schenk A, Kieven M, Holthaus M, Lehmann J, Lovenich L, et al. Physiol (1985) (2004) 97(2):491–8. doi:10.1152/japplphysiol.01404.2003
Exercise induced alterations in NK-cell cytotoxicity – methodological issues 147. Schuler PB, Leblanc PA, Marzilli TS. Effect of physical activity on the
and future perspectives. Exerc Immunol Rev (2017) 23:66–81. production of specific antibody in response to the 1998–99 influenza virus
129. McFarlin BK, Venable AS, Carpenter CK, Henning AL, Stephan O. Oral vaccine in older adults. J Sports Med Phys Fitness (2003) 43(3):404.
supplementation with baker’s yeast beta glucan is associated with altered 148. Grant RW, Mariani RA, Vieira VJ, Fleshner M, Smith TP, Keylock KT,
monocytes, T  cells and cytokines following a bout of strenuous exercise. et  al. Cardiovascular exercise intervention improves the primary antibody
Front Physiol (2017) 8:786. doi:10.3389/fphys.2017.00786 response to keyhole limpet hemocyanin (KLH) in previously sedentary
130. Witard OC, Turner JE, Jackman SR, Kies AK, Jeukendrup AE, Bosch JA, older adults. Brain Behav Immun (2008) 22(6):923–32. doi:10.1016/j.bbi.
et  al. High dietary protein restores overreaching induced impairments in 2008.01.006
leukocyte trafficking and reduces the incidence of upper respiratory tract 149. Woods JA, Keylock KT, Lowder T, Vieira VJ, Zelkovich W, Dumich S, et al.
infection in elite cyclists. Brain Behav Immun (2014) 39:211–9. doi:10.1016/j. Cardiovascular exercise training extends influenza vaccine seroprotection in
bbi.2013.10.002 sedentary older adults: the immune function intervention trial. J Am Geriatr
131. Albers R, Bourdet-Sicard R, Braun D, Calder PC, Herz U, Lambert C, et al. Soc (2009) 57(12):2183–91. doi:10.1111/j.1532-5415.2009.02563.x
Monitoring immune modulation by nutrition in the general population: 150. Yang Y, Verkuilen J, Rosengren KS, Mariani RA, Reed M, Grubisich SA,
identifying and substantiating effects on human health. Br J Nutr (2013) et  al. Effects of a traditional Taiji/Qigong curriculum on older adults’
110(Suppl 2):S1–30. doi:10.1017/S0007114513001505 immune response to influenza vaccine. Med Sport Sci (2008) 52:64–76.
132. Burns VE, Gallagher S. Antibody response to vaccination as a marker of doi:10.1159/000134285
in vivo immune function in psychophysiological research. Neurosci Biobehav 151. de Araujo AL, Silva LC, Fernandes JR, Matias Mde S, Boas LS, Machado CM,
Rev (2010) 35(1):122–6. doi:10.1016/j.neubiorev.2010.01.005 et al. Elderly men with moderate and intense training lifestyle present sus-
133. Edwards KM, Burns VE, Reynolds T, Carroll D, Drayson M, Ring C. tained higher antibody responses to influenza vaccine. Age (Dordr) (2015)
Acute stress exposure prior to influenza vaccination enhances antibody 37(6):105. doi:10.1007/s11357-015-9843-4
response in women. Brain Behav Immun (2006) 20(2):159–68. doi:10.1016/j. 152. Kapasi ZF, Catlin PA, Joyner DR, Lewis ML, Schwartz AL, Townsend EL.
bbi.2005.07.001 The effects of intense physical exercise on secondary antibody response in
134. Edwards KM, Burns VE, Allen LM, McPhee JS, Bosch JA, Carroll D, et al. young and old mice. Phys Ther (2000) 80(11):1076–86.
Eccentric exercise as an adjuvant to influenza vaccination in humans. Brain 153. Grande AJ, Reid H, Thomas EE, Nunan D, Foster C. Exercise prior to
Behav Immun (2007) 21(2):209–17. doi:10.1016/j.bbi.2006.04.158 influenza vaccination for limiting influenza incidence and its related
135. Edwards KM, Burns VE, Adkins AE, Carroll D, Drayson M, Ring C. complications in adults. Cochrane Database Syst Rev (2016) (8):CD011857.
Meningococcal A vaccination response is enhanced by acute stress in men. doi:10.1002/14651858.CD011857.pub2
Psychosom Med (2008) 70(2):147–51. doi:10.1097/PSY.0b013e318164232e 154. Warren K, Thompson N, Wannemuehler M, Kohut M. Antibody and CD8+
136. Edwards KM, Campbell JP, Ring C, Drayson MT, Bosch JA, Downes C, T  cell memory response to influenza A/PR/8/34 infection is reduced in
et al. Exercise intensity does not influence the efficacy of eccentric exercise treadmill-exercised mice, yet still protective. J Appl Physiol (1985) (2013)
as a behavioural adjuvant to vaccination. Brain Behav Immun (2010) 114(10):1413–20. doi:10.1152/japplphysiol.01355.2012
24(4):623–30. doi:10.1016/j.bbi.2010.01.009 155. Sim YJ, Yu S, Yoon KJ, Loiacono CM, Kohut ML. Chronic exercise reduces
137. Edwards KM, Pung MA, Tomfohr LM, Ziegler MG, Campbell JP, Drayson MT, illness severity, decreases viral load, and results in greater anti-inflammatory
et  al. Acute exercise enhancement of pneumococcal vaccination response: effects than acute exercise during influenza infection. J Infect Dis (2009)
a randomised controlled trial of weaker and stronger immune response. 200(9):1434–42. doi:10.1086/606014
Vaccine (2012) 30(45):6389–95. doi:10.1016/j.vaccine.2012.08.022 156. Lowder T, Padgett DA, Woods JA. Moderate exercise protects mice from
138. Ranadive SM, Cook M, Kappus RM, Yan H, Lane AD, Woods JA, et  al. death due to influenza virus. Brain Behav Immun (2005) 19(5):377–80.
Effect of acute aerobic exercise on vaccine efficacy in older adults. Med Sci doi:10.1016/j.bbi.2005.04.002
Sports Exerc (2014) 46(3):455–61. doi:10.1249/MSS.0b013e3182a75ff2 157. Nosbaum A, Vocanson M, Rozieres A, Hennino A, Nicolas JF. Allergic and
139. Long JE, Ring C, Drayson M, Bosch J, Campbell JP, Bhabra J, et al. Vaccination irritant contact dermatitis. Eur J Dermatol (2009) 19(4):325–32. doi:10.1684/
response following aerobic exercise: can a brisk walk enhance antibody ejd.2009.0686
response to pneumococcal and influenza vaccinations? Brain Behav Immun 158. Gulati N, Suarez-Farinas M, Fuentes-Duculan J, Gilleaudeau P, Sullivan-
(2012) 26(4):680–7. doi:10.1016/j.bbi.2012.02.004 Whalen M, Correa da Rosa J, et  al. Molecular characterization of human
140. Edwards KM, Pascoe AR, Fiatarone-Singh MA, Singh NA, Kok J, Booy R. skin response to diphencyprone at peak and resolution phases: therapeutic
A randomised controlled trial of resistance exercise prior to administration insights. J Invest Dermatol (2014) 134(10):2531–40. doi:10.1038/jid.2014.196

Frontiers in Immunology  |  www.frontiersin.org 18 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

159. Diment BC, Fortes MB, Edwards JP, Hanstock HG, Ward MD, Dunstall HM, adipose tissue macrophages independently of classic activation. Cell Metab
et al. Exercise intensity and duration effects on in vivo immunity. Med Sci (2013) 18(6):816–30. doi:10.1016/j.cmet.2013.11.001
Sports Exerc (2015) 47(7):1390–8. doi:10.1249/MSS.0000000000000562 180. Chavez-Galan L, Olleros ML, Vesin D, Garcia I. Much more than M1 and
160. Harper Smith AD, Coakley SL, Ward MD, Macfarlane AW, Friedmann PS, M2 macrophages, there are also CD169(+) and TCR(+) macrophages. Front
Walsh NP. Exercise-induced stress inhibits both the induction and eli­ Immunol (2015) 6:263. doi:10.3389/fimmu.2015.00263
citation phases of in vivo T-cell-mediated immune responses in humans. 181. Mills CD. Anatomy of a discovery: m1 and m2 macrophages. Front Immunol
Brain Behav Immun (2011) 25(6):1136–42. doi:10.1016/j.bbi.2011. (2015) 6:212. doi:10.3389/fimmu.2015.00212
02.014 182. Bueno V, Sant’Anna OA, Lord JM. Ageing and myeloid-derived suppressor
161. Ceddia MA, Price EA, Kohlmeier CK, Evans JK, Lu Q, McAuley E, et  al. cells: possible involvement in immunosenescence and age-related disease.
Differential leukocytosis and lymphocyte mitogenic response to acute maxi- Age (Dordr) (2014) 36(6):9729. doi:10.1007/s11357-014-9729-x
mal exercise in the young and old. Med Sci Sports Exerc (1999) 31(6):829–36. 183. Shearer GM. Th1/Th2 changes in aging. Mech Ageing Dev (1997) 94(1–3):1–5.
doi:10.1097/00005768-199906000-00011 184. Schmitt V, Rink L, Uciechowski P. The Th17/Treg balance is disturbed during
162. Woods JA, Evans JK, Wolters BW, Ceddia MA, McAuley E. Effects of max- aging. Exp Gerontol (2013) 48(12):1379–86. doi:10.1016/j.exger.2013.
imal exercise on natural killer (NK) cell cytotoxicity and responsiveness to 09.003
interferon-alpha in the young and old. J Gerontol A Biol Sci Med Sci (1998) 185. Fulop T, Larbi A, Pawelec G. Human T  cell aging and the impact of
53(6):B430–7. doi:10.1093/gerona/53A.6.B430 persistent viral infections. Front Immunol (2013) 4:271. doi:10.3389/
163. Simpson RJ, Lowder TW, Spielmann G, Bigley AB, LaVoy EC, Kunz H. fimmu.2013.00271
Exercise and the aging immune system. Ageing Res Rev (2012) 11(3):404–20. 186. Pawelec G. Hallmarks of human “immunosenescence”: adaptation or dysreg-
doi:10.1016/j.arr.2012.03.003 ulation? Immun Ageing (2012) 9(1):15. doi:10.1186/1742-4933-9-15
164. Kohut ML, Senchina DS. Reversing age-associated immunosenescence via 187. Geiger H, de Haan G, Florian MC. The ageing haematopoietic stem
exercise. Exerc Immunol Rev (2004) 10:6–41. cell compartment. Nat Rev Immunol (2013) 13(5):376–89. doi:10.1038/
165. Woods JA, Lowder TW, Keylock KT. Can exercise training improve immune nri3433
function in the aged? Ann N Y Acad Sci (2002) 959:117–27. doi:10.1111/j. 188. Roux A, Mourin G, Larsen M, Fastenackels S, Urrutia A, Gorochov G, et al.
1749-6632.2002.tb02088.x Differential impact of age and cytomegalovirus infection on the gamma­
166. Simpson RJ, Bigley AB, Spielmann G, LaVoy EC, Kunz H, Bollard CM. delta T  cell compartment. J Immunol (2013) 191(3):1300–6. doi:10.4049/
Human cytomegalovirus infection and the immune response to exercise. jimmunol.1202940
Exerc Immunol Rev (2016) 22:8–27. 189. Colonna-Romano G, Aquino A, Bulati M, Lio D, Candore G, Oddo G, et al.
167. Bartlett DB, Fox O, McNulty CL, Greenwood HL, Murphy L, Sapey E, et al. Impairment of gamma/delta T  lymphocytes in elderly: implications for
Habitual physical activity is associated with the maintenance of neutrophil immunosenescence. Exp Gerontol (2004) 39(10):1439–46. doi:10.1016/j.
migratory dynamics in healthy older adults. Brain Behav Immun (2016) exger.2004.07.005
56:12–20. doi:10.1016/j.bbi.2016.02.024 190. Michishita Y, Hirokawa M, Guo YM, Abe Y, Liu J, Ubukawa K, et  al.
168. Bartlett DB, Shepherd SO, Wilson OJ, Adlan AM, Wagenmakers AJM, Shaw CS, Age-associated alteration of gammadelta T-cell repertoire and different pro-
et al. Neutrophil and monocyte bactericidal responses to 10 weeks of low- files of activation-induced death of Vdelta1 and Vdelta2 T cells. Int J Hematol
volume high-intensity interval or moderate-intensity continuous training in (2011) 94(3):230–40. doi:10.1007/s12185-011-0907-7
sedentary adults. Oxid Med Cell Longev (2017) 2017:8148742. doi:10.1155/ 191. Jagger A, Shimojima Y, Goronzy JJ, Weyand CM. Regulatory T  cells and
2017/8148742 the immune aging process: a mini-review. Gerontology (2014) 60(2):130–7.
169. Shephard RJ. Development of the discipline of exercise immunology. Exerc doi:10.1159/000355303
Immunol Rev (2010) 16:194–222. 192. Siegrist CA, Aspinall R. B-cell responses to vaccination at the extremes
170. Hazeldine J, Lord JM. Innate immunesenescence: underlying mechanisms of age. Nat Rev Immunol (2009) 9(3):185–94. doi:10.1038/nri2508
and clinical relevance. Biogerontology (2015) 16(2):187–201. doi:10.1007/ 193. Appay V, Sauce D. Naive T  cells: the crux of cellular immune aging? Exp
s10522-014-9514-3 Gerontol (2014) 54:90–3. doi:10.1016/j.exger.2014.01.003
171. Peralbo E, Alonso C, Solana R. Invariant NKT and NKT-like lymphocytes: 194. Pawelec G. Immunosenenescence: role of cytomegalovirus. Exp Gerontol
two different T  cell subsets that are differentially affected by ageing. Exp (2013) 54:1–5. doi:10.1016/j.exger.2013.11.010
Gerontol (2007) 42(8):703–8. doi:10.1016/j.exger.2007.05.002 195. Wikby A, Maxson P, Olsson J, Johansson B, Ferguson FG. Changes in CD8
172. Ziegler-Heitbrock L, Ancuta P, Crowe S, Dalod M, Grau V, Hart DN, et al. and CD4 lymphocyte subsets, T  cell proliferation responses and non-
Nomenclature of monocytes and dendritic cells in blood. Blood (2010) survival in the very old: the Swedish longitudinal OCTO-immune study. Mech
116(16):e74–80. doi:10.1182/blood-2010-02-258558 Ageing Dev (1998) 102(2–3):187–98. doi:10.1016/S0047-6374(97)00151-6
173. Goh J, Ladiges WC. Exercise enhances wound healing and prevents cancer 196. Pawelec G, Ferguson FG, Wikby A. The SENIEUR protocol after 16 years.
progression during aging by targeting macrophage polarity. Mech Ageing Dev Mech Ageing Dev (2001) 122(2):132–4. doi:10.1016/S0047-6374(00)00240-2
(2014) 139:41–8. doi:10.1016/j.mad.2014.06.004 197. Wikby A, Johansson B, Ferguson F, Olsson J. Age-related changes in immune
174. Kanneganti TD, Dixit VD. Immunological complications of obesity. Nat parameters in a very old population of Swedish people: a longitudinal study.
Immunol (2012) 13(8):707–12. doi:10.1038/ni.2343 Exp Gerontol (1994) 29(5):531–41. doi:10.1016/0531-5565(94)90036-1
175. Garg SK, Delaney C, Shi H, Yung R. Changes in adipose tissue macrophages 198. Wikby A, Nilsson BO, Forsey R, Thompson J, Strindhall J, Lofgren S, et al.
and T cells during aging. Crit Rev Immunol (2014) 34(1):1–14. doi:10.1615/ The immune risk phenotype is associated with IL-6 in the terminal decline
CritRevImmunol.2013006833 stage: findings from the Swedish NONA immune longitudinal study of very
176. Kratz M, Coats BR, Hisert KB, Hagman D, Mutskov V, Peris E, et  al. late life functioning. Mech Ageing Dev (2006) 127(8):695–704. doi:10.1016/j.
Meta­bolic dysfunction drives a mechanistically distinct proinflammatory mad.2006.04.003
phenotype in adipose tissue macrophages. Cell Metab (2014) 20(4):614–25. 199. Wikby A, Ferguson F, Forsey R, Thompson J, Strindhall J, Lofgren S, et al.
doi:10.1016/j.cmet.2014.08.010 An immune risk phenotype, cognitive impairment, and survival in very late
177. Bourlier V, Zakaroff-Girard A, Miranville A, De Barros S, Maumus M, life: impact of allostatic load in Swedish octogenarian and nonagenarian
Sengenes C, et al. Remodeling phenotype of human subcutaneous adipose humans. J Gerontol A Biol Sci Med Sci (2005) 60(5):556–65. doi:10.1093/
tissue macrophages. Circulation (2008) 117(6):806–15. doi:10.1161/ gerona/60.5.556
CIRCULATIONAHA.107.724096 200. Olsson J, Wikby A, Johansson B, Lofgren S, Nilsson BO, Ferguson FG. Age-related
178. Zeyda M, Farmer D, Todoric J, Aszmann O, Speiser M, Gyori G, et  al. change in peripheral blood T-lymphocyte subpopulations and cytomegalovirus
Human adipose tissue macrophages are of an anti-inflammatory phenotype infection in the very old: the Swedish longitudinal OCTO immune study. Mech
but capable of excessive pro-inflammatory mediator production. Int J Obes Ageing Dev (2000) 121(1–3):187–201. doi:10.1016/S0047-6374(00)00210-4
(Lond) (2007) 31(9):1420–8. doi:10.1038/sj.ijo.0803632 201. Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M, Ottaviani E, et al.
179. Xu X, Grijalva A, Skowronski A, van Eijk M, Serlie MJ, Ferrante AW Jr. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann
Obesity activates a program of lysosomal-dependent lipid metabolism in N Y Acad Sci (2000) 908:244–54. doi:10.1111/j.1749-6632.2000.tb06651.x

Frontiers in Immunology  |  www.frontiersin.org 19 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

202. Franceschi C, Capri M, Monti D, Giunta S, Olivieri F, Sevini F, et  al. 220. Shimizu K, Kimura F, Akimoto T, Akama T, Tanabe K, Nishijima T, et al.
Inflammaging and anti-inflammaging: a systemic perspective on aging Effect of moderate exercise training on T-helper cell subpopulations in
and longevity emerged from studies in humans. Mech Ageing Dev (2007) elderly people. Exerc Immunol Rev (2008) 14:24–37.
128(1):92–105. doi:10.1016/j.mad.2006.11.016 221. Cao Dinh H, Beyer I, Mets T, Onyema OO, Njemini R, Renmans W, et al. Effects
203. Giovannini S, Onder G, Liperoti R, Russo A, Carter C, Capoluongo E, of physical exercise on markers of cellular immunosenescence: a systematic
et al. Interleukin-6, C-reactive protein, and tumor necrosis factor-alpha as review. Calcif Tissue Int (2017) 100(2):193–215. doi:10.1007/s00223-016-0212-9
pre­dictors of mortality in frail, community-living elderly individuals. J Am 222. Kruger K, Frost S, Most E, Volker K, Pallauf J, Mooren FC. Exercise affects
Geriatr Soc (2011) 59(9):1679–85. doi:10.1111/j.1532-5415.2011.03570.x tissue lymphocyte apoptosis via redox-sensitive and Fas-dependent signaling
204. Baune BT, Rothermundt M, Ladwig KH, Meisinger C, Berger K. Systemic pathways. Am J Physiol Regul Integr Comp Physiol (2009) 296(5):R1518–27.
inflammation (interleukin 6) predicts all-cause mortality in men: results doi:10.1152/ajpregu.90994.2008
from a 9-year follow-up of the MEMO Study. Age (Dordr) (2011) 33(2): 223. Khan N, Shariff N, Cobbold M, Bruton R, Ainsworth JA, Sinclair AJ,
209–17. doi:10.1007/s11357-010-9165-5 et  al. Cytomegalovirus seropositivity drives the CD8 T  cell repertoire
205. Bruunsgaard H, Ladelund S, Pedersen AN, Schroll M, Jorgensen T, toward greater clonality in healthy elderly individuals. J Immunol (2002)
Pedersen BK. Predicting death from tumour necrosis factor-alpha and 169(4):1984–92. doi:10.4049/jimmunol.169.4.1984
interleukin-6 in 80-year-old people. Clin Exp Immunol (2003) 132(1):24–31. 224. Mueller YM, De Rosa SC, Hutton JA, Witek J, Roederer M, Altman JD, et al.
doi:10.1046/j.1365-2249.2003.02137.x Increased CD95/Fas-induced apoptosis of HIV-specific CD8(+) T  cells.
206. McElhaney JE, Kuchel GA, Zhou X, Swain SL, Haynes L. T-cell immunity Immunity (2001) 15(6):871–82. doi:10.1016/S1074-7613(01)00246-1
to influenza in older adults: a pathophysiological framework for develop- 225. Wang JS, Lin CT. Systemic hypoxia promotes lymphocyte apoptosis induced
ment of more effective vaccines. Front Immunol (2016) 7:41. doi:10.3389/ by oxidative stress during moderate exercise. Eur J Appl Physiol (2010)
fimmu.2016.00041 108(2):371–82. doi:10.1007/s00421-009-1231-2
207. Frasca D, Blomberg BB. Aging, cytomegalovirus (CMV) and influenza vac- 226. Takahashi A, Hanson MG, Norell HR, Havelka AM, Kono K, Malmberg KJ,
cine responses. Hum Vaccin Immunother (2016) 12(3):682–90. doi:10.1080/ et  al. Preferential cell death of CD8+ effector memory (CCR7-CD45RA-)
21645515.2015.1105413 T  cells by hydrogen peroxide-induced oxidative stress. J Immunol (2005)
208. Brown FF, Bigley AB, Sherry C, Neal CM, Witard OC, Simpson RJ, et  al. 174(10):6080–7. doi:10.4049/jimmunol.174.10.6080
Training status and sex influence on senescent T-lymphocyte redistribution 227. Stocchi R, Ward KN, Fanin R, Baccarani M, Apperley JF. Management of
in response to acute maximal exercise. Brain Behav Immun (2014) 39:152–9. human cytomegalovirus infection and disease after allogeneic bone marrow
doi:10.1016/j.bbi.2013.10.031 transplantation. Haematologica (1999) 84(1):71–9.
209. Brown FF, Bigley AB, Ross JC, LaVoy EC, Simpson RJ, Galloway SD. 228. Derhovanessian E, Maier AB, Hahnel K, Zelba H, de Craen AJ, Roelofs H,
T-lymphocyte populations following a period of high volume training in et al. Lower proportion of naive peripheral CD8+ T cells and an unopposed
female soccer players. Physiol Behav (2015) 152(Pt A):175–81. doi:10.1016/j. pro-inflammatory response to human cytomegalovirus proteins in  vitro
physbeh.2015.09.027 are associated with longer survival in very elderly people. Age (Dordr)
210. Gustafson MP, DiCostanzo AC, Wheatley CM, Kim CH, Bornschlegl S, (2013) 35(4):1387–99. doi:10.1007/s11357-012-9425-7
Gastineau DA, et al. A systems biology approach to investigating the influ- 229. Sylwester AW, Mitchell BL, Edgar JB, Taormina C, Pelte C, Ruchti F, et al.
ence of exercise and fitness on the composition of leukocytes in peripheral Broadly targeted human cytomegalovirus-specific CD4+ and CD8+ T cells
blood. J Immunother Cancer (2017) 5:30. doi:10.1186/s40425-017-0231-8 dominate the memory compartments of exposed subjects. J Exp Med (2005)
211. Prieto-Hinojosa A, Knight A, Compton C, Gleeson M, Travers PJ. Redu­ 202(5):673–85. doi:10.1084/jem.20050882
ced thymic output in elite athletes. Brain Behav Immun (2014) 39:75–9. 230. Franceschi C, Bonafe M, Valensin S. Human immunosenescence: the
doi:10.1016/j.bbi.2014.01.004 prevailing of innate immunity, the failing of clonotypic immunity, and the
212. Moro-Garcia MA, Fernandez-Garcia B, Echeverria A, Rodriguez-Alonso M, filling of immunological space. Vaccine (2000) 18(16):1717–20. doi:10.1016/
Suarez-Garcia FM, Solano-Jaurrieta JJ, et al. Frequent participation in high S0264-410X(99)00513-7
volume exercise throughout life is associated with a more differentiated adap- 231. Akbar AN, Fletcher JM. Memory T cell homeostasis and senescence during
tive immune response. Brain Behav Immun (2014) 39:61–74. doi:10.1016/j. aging. Curr Opin Immunol (2005) 17(5):480–5. doi:10.1016/j.coi.2005.07.019
bbi.2013.12.014 232. van Leeuwen EM, Koning JJ, Remmerswaal EB, van Baarle D, van Lier RA,
213. Teixeira AM, Rama L, Carvalho HM, Borges G, Carvalheiro T, Gleeson M, ten Berge IJ. Differential usage of cellular niches by cytomegalovirus versus
et  al. Changes in naive and memory T-cells in elite swimmers during a EBV- and influenza virus-specific CD8+ T cells. J Immunol (2006) 177(8):
winter training season. Brain Behav Immun (2014) 39:186–93. doi:10.1016/j. 4998–5005. doi:10.4049/jimmunol.177.8.4998
bbi.2014.01.002 233. Huster KM, Stemberger C, Gasteiger G, Kastenmuller W, Drexler I, Busch DH.
214. Cosgrove C, Galloway SD, Neal C, Hunter AM, McFarlin BK, Spielmann G, Cutting edge: memory CD8 T cell compartment grows in size with immu-
et al. The impact of 6-month training preparation for an Ironman triathlon nological experience but nevertheless can lose function. J Immunol (2009)
on the proportions of naive, memory and senescent T cells in resting blood. 183(11):6898–902. doi:10.4049/jimmunol.0902454
Eur J Appl Physiol (2012) 112(8):2989–98. doi:10.1007/s00421-011-2273-9 234. Ferrando-Martinez S, Franco JM, Ruiz-Mateos E, Hernandez A, Ordonez A,
215. Dopico XC, Evangelou M, Ferreira RC, Guo H, Pekalski ML, Smyth DJ, Gutierrez E, et al. A reliable and simplified sj/beta-TREC ratio quantification
et  al. Widespread seasonal gene expression reveals annual differences in method for human thymic output measurement. J Immunol Methods (2010)
human immunity and physiology. Nat Commun (2015) 6:7000. doi:10.1038/ 352(1–2):111–7. doi:10.1016/j.jim.2009.11.007
ncomms8000 235. Radtke F, MacDonald HR, Tacchini-Cottier F. Regulation of innate and adap-
216. Goldinger A, Shakhbazov K, Henders AK, McRae AF, Montgomery GW, tive immunity by Notch. Nat Rev Immunol (2013) 13(6):427–37. doi:10.1038/
Powell JE. Seasonal effects on gene expression. PLoS One (2015) 10(5): nri3445
e0126995. doi:10.1371/journal.pone.0126995 236. Haugen F, Norheim F, Lian H, Wensaas AJ, Dueland S, Berg O, et  al.
217. Spielmann G, McFarlin BK, O’Connor DP, Smith PJ, Pircher H, Simpson RJ.  IL-7 is expressed and secreted by human skeletal muscle cells. Am J Physiol
Aerobic fitness is associated with lower proportions of senescent blood T-cells in Cell Physiol (2010) 298(4):C807–16. doi:10.1152/ajpcell.00094.2009
man. Brain Behav Immun (2011) 25(8):1521–9. doi:10.1016/j.bbi.2011.07.226 237. Fry TJ, Mackall CL. The many faces of IL-7: from lymphopoiesis to periph-
218. Silva LC, de Araujo AL, Fernandes JR, Matias Mde S, Silva PR, Duarte AJ, eral T  cell maintenance. J Immunol (2005) 174(11):6571–6. doi:10.4049/
et al. Moderate and intense exercise lifestyles attenuate the effects of aging on jimmunol.174.11.6571
telomere length and the survival and composition of T cell subpopulations. 238. Tchernof A, Despres JP. Pathophysiology of human visceral obesity: an
Age (Dordr) (2016) 38(1):24. doi:10.1007/s11357-016-9879-0 update. Physiol Rev (2013) 93(1):359–404. doi:10.1152/physrev.00033.2011
219. Duggal NA, Pollock RD, Lazarus NR, Harridge S, Lord JM. Major features of 239. Trim W, Turner JE, Thompson D. Parallels in immunometabolic adipose
immunesenescence, including reduced thymic output, are ameliorated by high tissue dysfunction with ageing and obesity. Front Immunol (2018) 9:169.
levels of physiacl activity in adulthood. Aging Cell (2018). doi:10.1111/acel.12750 doi:10.3389/fimmu.2018.00169

Frontiers in Immunology  |  www.frontiersin.org 20 April 2018 | Volume 9 | Article 648


Campbell and Turner Exercise, Health, and Immunity Across the Lifespan

240. Nieman DC, Henson DA, Nehlsen-Cannarella SL, Ekkens M, Utter AC, 247. Gleeson M, Bishop NC, Stensel DJ, Lindley MR, Mastana SS, Nimmo MA.
Butterworth DE, et al. Influence of obesity on immune function. J Am Diet The anti-inflammatory effects of exercise: mechanisms and implications
Assoc (1999) 99(3):294–9. doi:10.1016/S0002-8223(99)00077-2 for the prevention and treatment of disease. Nat Rev Immunol (2011) 11(9):
241. Muezzinler A, Zaineddin AK, Brenner H. Body mass index and leukocyte 607–15. doi:10.1038/nri3041
telomere length in adults: a systematic review and meta-analysis. Obes Rev 248. Radak Z, Chung HY, Goto S. Systemic adaptation to oxidative challenge
(2014) 15(3):192–201. doi:10.1111/obr.12126 induced by regular exercise. Free Radic Biol Med (2008) 44(2):153–9.
242. van der Weerd K, Dik WA, Schrijver B, Schweitzer DH, Langerak AW, doi:10.1016/j.freeradbiomed.2007.01.029
Drexhage HA, et al. Morbidly obese human subjects have increased periph- 249. Henson SM, Lanna A, Riddell NE, Franzese O, Macaulay R, Griffiths SJ,
eral blood CD4+ T cells with skewing toward a Treg- and Th2-dominated et al. p38 signaling inhibits mTORC1-independent autophagy in senescent
phenotype. Diabetes (2012) 61(2):401–8. doi:10.2337/db11-1065 human CD8(+) T cells. J Clin Invest (2014) 124(9):4004–16. doi:10.1172/
243. Alam I, Larbi A, Pawelec G. Nutritional status influences peripheral immune JCI75051
cell phenotypes in healthy men in rural Pakistan. Immun Ageing (2012)
9(1):16. doi:10.1186/1742-4933-9-16 Conflict of Interest Statement: The authors declare that the research was con-
244. Spielmann G, Johnston CA, O’Connor DP, Foreyt JP, Simpson RJ. Excess ducted in the absence of any commercial or financial relationships that could be
body mass is associated with T  cell differentiation indicative of immune construed as a potential conflict of interest.
ageing in children. Clin Exp Immunol (2014) 176(2):246–54. doi:10.1111/cei.
12267 Copyright © 2018 Campbell and Turner. This is an open-access article distri­buted
245. Costanzo AE, Taylor KR, Dutt S, Han PP, Fujioka K, Jameson JM. Obesity under the terms of the Creative Commons Attribution License (CC BY). The use,
impairs gammadelta T cell homeostasis and antiviral function in humans. distribution or reproduction in other forums is permitted, provided the original
PLoS One (2015) 10(3):e0120918. doi:10.1371/journal.pone.0120918 author(s) and the copyright owner are credited and that the original publication in
246. Aroor AR, DeMarco VG. Oxidative stress and obesity: the chicken or the egg? this journal is cited, in accordance with accepted academic practice. No use, distribu-
Diabetes (2014) 63(7):2216–8. doi:10.2337/db14-0424 tion or reproduction is permitted which does not comply with these terms.

Frontiers in Immunology  |  www.frontiersin.org 21 April 2018 | Volume 9 | Article 648

You might also like